1
|
Ding JY, Meng TT, Du RL, Song XB, Li YX, Gao J, Ji R, He QY. Bibliometrics of trends in global research on the roles of stem cells in myocardial fibrosis therapy. World J Stem Cells 2024; 16:1086-1105. [PMID: 39734477 PMCID: PMC11669986 DOI: 10.4252/wjsc.v16.i12.1086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 10/05/2024] [Accepted: 11/11/2024] [Indexed: 12/13/2024] Open
Abstract
BACKGROUND Myocardial fibrosis, a condition linked to several cardiovascular diseases, is associated with a poor prognosis. Stem cell therapy has emerged as a potential treatment option and the application of stem cell therapy has been studied extensively. However, a comprehensive bibliometric analysis of these studies has yet to be conducted. AIM To map thematic trends, analyze research hotspots, and project future directions of stem cell-based myocardial fibrosis therapy. METHODS We conducted a bibliometric and visual analysis of studies in the Web of Science Core Collection using VOSviewer and Microsoft Excel. The dataset included 1510 articles published between 2001 and 2024. Countries, organizations, authors, references, keywords, and co-citation networks were examined to identify evolving research trends. RESULTS Our findings revealed a steady increase in the number of publications, with a projected increase to over 200 publications annually by 2030. Initial research focused on stem cell-based therapy, particularly for myocardial infarction and heart failure. More recently, there has been a shift toward cell-free therapy, involving extracellular vesicles, exosomes, and microRNAs. Key research topics include angiogenesis, inflammation, apoptosis, autophagy, and oxidative stress. CONCLUSION This analysis highlights the evolution of stem cell therapies for myocardial fibrosis, with emerging interest in cell-free approaches. These results are expected to guide future scientific exploration and decision-making.
Collapse
Affiliation(s)
- Jing-Yi Ding
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Tian-Tian Meng
- Department of Rehabilitation, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing 100071, China
| | - Ruo-Lin Du
- Department of Emergency Medicine, South Branch of Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Xin-Bin Song
- Department of Intensive Care Unit, Zhumadian Hospital of Traditional Chinese Medicine, Zhumadian 463000, Henan Province, China
| | - Yi-Xiang Li
- Department of Chinese Medicine, The Third People's Hospital of Henan Province, Zhengzhou 450000 Henan Province, China
| | - Jing Gao
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Ran Ji
- Department of Intensive Care Unit, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Qing-Yong He
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China.
| |
Collapse
|
2
|
Baccouche BM, Elde S, Wang H, Woo YJ. Structural, angiogenic, and immune responses influencing myocardial regeneration: a glimpse into the crucible. NPJ Regen Med 2024; 9:18. [PMID: 38688935 PMCID: PMC11061134 DOI: 10.1038/s41536-024-00357-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Accepted: 03/15/2024] [Indexed: 05/02/2024] Open
Abstract
Complete cardiac regeneration remains an elusive therapeutic goal. Although much attention has been focused on cardiomyocyte proliferation, especially in neonatal mammals, recent investigations have unearthed mechanisms by which non-cardiomyocytes, such as endothelial cells, fibroblasts, macrophages, and other immune cells, play critical roles in modulating the regenerative capacity of the injured heart. The degree to which each of these cell types influence cardiac regeneration, however, remains incompletely understood. This review highlights the roles of these non-cardiomyocytes and their respective contributions to cardiac regeneration, with emphasis on natural heart regeneration after cardiac injury during the neonatal period.
Collapse
Affiliation(s)
- Basil M Baccouche
- Stanford University Department of Cardiothoracic Surgery, Palo Alto, CA, USA
| | - Stefan Elde
- Stanford University Department of Cardiothoracic Surgery, Palo Alto, CA, USA
| | - Hanjay Wang
- Stanford University Department of Cardiothoracic Surgery, Palo Alto, CA, USA
| | - Y Joseph Woo
- Stanford University Department of Cardiothoracic Surgery, Palo Alto, CA, USA.
| |
Collapse
|
3
|
Soetisna TW, Thamrin AMH, Permadijana D, Ramadhani ANE, Santoso A, Mansyur M. Intramyocardial Stem Cell Transplantation during Coronary Artery Bypass Surgery Safely Improves Cardiac Function: Meta-Analysis of 20 Randomized Clinical Trials. J Clin Med 2023; 12:4430. [PMID: 37445467 DOI: 10.3390/jcm12134430] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 06/26/2023] [Accepted: 06/28/2023] [Indexed: 07/15/2023] Open
Abstract
IMSC transplantation during CABG is considered one of the most promising methods to effectively deliver stem cells and has been widely studied in many trials. But the results of outcomes and safety of this modality still vary widely. We conducted this meta-analysis of randomized controlled trials (RCTs) to evaluate not only the outcome but also the safety of this promising method. A meta-analysis was performed according to Preferred Reporting Items for Systematic Reviews and Meta-Analysis (PRISMA) guidelines. A comprehensive literature search was undertaken using the PubMed, Scopus, and Cochrane databases. Articles were thoroughly evaluated and analyzed. Twenty publications about IMSC during CABG were included. Primary outcomes were measured using LVEF, LVESV, LVESVI, LVESD, LVEDV, LVEDVI, LVEDD, WMSI, and 6-MWT. Safety measures were depicted by total deaths, MACE, CRD, CVA, myocardial infarction, ventricular arrhythmia, and cardiac-related readmission. IMSC transplantation during CABG significantly improved LVEF (MD = 3.89%; 95% CI = 1.31% to 6.46%; p = 0.003) and WMSI (MD = 0.28; 95% CI = 0.01-0.56; p = 0.04). Most of the other outcomes showed favorable results for the IMSC group but were not statistically significant. The safety analysis also showed no significant risk difference for IMSC transplantation compared to CABG alone. IMSC during CABG can safely improve cardiac function and tend to improve cardiac volumes and dimensions. The analysis and application of influencing factors that increase patients' responses to IMSC transplantation are important to achieve long-term improvement.
Collapse
Affiliation(s)
- Tri Wisesa Soetisna
- Adult Cardiac Surgery Division, Department of Thoracic and Cardiovascular Surgery, Harapan Kita National Cardiovascular Center Hospital, Jakarta 11420, Indonesia
- Department of Thoracic and Cardiovascular Surgery, Faculty of Medicine, University of Indonesia, Jakarta 10430, Indonesia
| | - Ahmad Muslim Hidayat Thamrin
- Adult Cardiac Surgery Division, Department of Thoracic and Cardiovascular Surgery, Harapan Kita National Cardiovascular Center Hospital, Jakarta 11420, Indonesia
- Faculty of Medicine, Syarif Hidayatullah State Islamic University, Haji Hospital, Jakarta 13560, Indonesia
| | - Diajeng Permadijana
- Adult Cardiac Surgery Division, Department of Thoracic and Cardiovascular Surgery, Harapan Kita National Cardiovascular Center Hospital, Jakarta 11420, Indonesia
| | - Andi Nurul Erisya Ramadhani
- Adult Cardiac Surgery Division, Department of Thoracic and Cardiovascular Surgery, Harapan Kita National Cardiovascular Center Hospital, Jakarta 11420, Indonesia
| | - Anwar Santoso
- Department of Cardiology and Vascular Medicine, Harapan Kita National Cardiovascular Center Hospital, Jakarta 11420, Indonesia
- Department of Cardiology and Vascular Medicine, Faculty of Medicine, University of Indonesia, Jakarta 10430, Indonesia
| | - Muchtaruddin Mansyur
- Department of Community Medicine, Faculty of Medicine, University of Indonesia, Jakarta 10430, Indonesia
| |
Collapse
|
4
|
Song J, He K, Hou J. Autologous bone marrow stem cell transplantation for patients undergoing coronary artery bypass grafting: a meta-analysis of 22 randomized controlled trials. J Cardiothorac Surg 2022; 17:167. [PMID: 35752847 PMCID: PMC9233763 DOI: 10.1186/s13019-022-01838-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 04/17/2022] [Indexed: 02/08/2023] Open
Abstract
Background Although the safety and feasibility of coronary artery bypass grafting (CABG) and bone marrow stem cell (BMSC) transplantation have been established, the effectiveness of this approach compared with CABG alone remains controversial. The aim of this updated meta-analysis of randomized controlled trials was to evaluate the efficacy of this procedure. Methods A random-effects meta-analysis was conducted using studies sourced from the PubMed, Embase, and Cochrane literature databases to compare patients who received isolated CABG (CABG group) and BMSC transplantation with CABG (BMSC group). 22 studies were included. Results A total of 22 relevant publications with 820 patients were included. 432 patients received BMSC transplantation with CABG and 388 patients received isolated CABG. Compared with the CABG group, the BMSC transplantation group exhibited an improvement in the left ventricular (LV) ejection fraction (mean difference (MD) = 3.87%; 95% confidence interval (CI): 1.93–5.80%; P < 0.001). Conclusion The present evidence suggests that autologous BMSC transplantation for patients undergoing CABG appears to be associated with an improvement in LV function compared with CABG alone. However, heterogeneity in the data suggests that patients respond differently to this therapy. Further research is needed to understand these differences. Supplementary Information The online version contains supplementary material available at 10.1186/s13019-022-01838-2.
Collapse
Affiliation(s)
- Juelin Song
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Kang He
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Jianglong Hou
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
5
|
Xu Z, Neuber S, Nazari-Shafti T, Liu Z, Dong F, Stamm C. Impact of procedural variability and study design quality on the efficacy of cell-based therapies for heart failure - a meta-analysis. PLoS One 2022; 17:e0261462. [PMID: 34986181 PMCID: PMC8730409 DOI: 10.1371/journal.pone.0261462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 12/02/2021] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND Cell-based therapy has long been considered a promising strategy for the treatment of heart failure (HF). However, its effectiveness in the clinical setting is now doubted. Because previous meta-analyses provided conflicting results, we sought to review all available data focusing on cell type and trial design. METHODS AND FINDINGS The electronic databases PubMed, Cochrane library, ClinicalTrials.gov, and EudraCT were searched for randomized controlled trials (RCTs) utilizing cell therapy for HF patients from January 1, 2000 to December 31, 2020. Forty-three RCTs with 2855 participants were identified. The quality of the reported study design was assessed by evaluating the risk-of-bias (ROB). Primary outcomes were defined as mortality rate and left ventricular ejection fraction (LVEF) change from baseline. Secondary outcomes included both heart function data and clinical symptoms/events. Between-study heterogeneity was assessed using the I2 index. Subgroup analysis was performed based on HF type, cell source, cell origin, cell type, cell processing, type of surgical intervention, cell delivery routes, cell dose, and follow-up duration. Only 10 of the 43 studies had a low ROB for all method- and outcome parameters. A higher ROB was associated with a greater increase in LVEF. Overall, there was no impact on mortality for up to 12 months follow-up, and a clinically irrelevant average LVEF increase by LVEF (2.4%, 95% CI = 0.75-4.05, p = 0.004). Freshly isolated, primary cells tended to produce better outcomes than cultured cell products, but there was no clear impact of the cell source tissue, bone marrow cell phenotype or cell chricdose (raw or normalized for CD34+ cells). A meaningful increase in LVEF was only observed when cell therapy was combined with myocardial revascularization. CONCLUSIONS The published results suggest a small increase in LVEF following cell therapy for heart failure, but publication bias and methodologic shortcomings need to be taken into account. Given that cardiac cell therapy has now been pursued for 20 years without real progress, further efforts should not be made. STUDY REGISTRY NUMBER This meta-analysis is registered at the international prospective register of systematic reviews, number CRD42019118872.
Collapse
Affiliation(s)
- Zhiyi Xu
- Berlin Institute of Health Center for Regenerative Therapies, Charité–Universitätsmedizin Berlin, Berlin, Germany
| | - Sebastian Neuber
- Berlin Institute of Health Center for Regenerative Therapies, Charité–Universitätsmedizin Berlin, Berlin, Germany
- Department of Cardiothoracic and Vascular Surgery, German Heart Center Berlin, Berlin, Germany
- German Centre for Cardiovascular Research, Partner Site Berlin, Berlin, Germany
| | - Timo Nazari-Shafti
- Berlin Institute of Health Center for Regenerative Therapies, Charité–Universitätsmedizin Berlin, Berlin, Germany
- Department of Cardiothoracic and Vascular Surgery, German Heart Center Berlin, Berlin, Germany
- German Centre for Cardiovascular Research, Partner Site Berlin, Berlin, Germany
- Berlin Institute of Health at Charité, Universitätsmedizin Berlin, Berlin, Germany
| | - Zihou Liu
- Berlin Institute of Health Center for Regenerative Therapies, Charité–Universitätsmedizin Berlin, Berlin, Germany
| | - Fengquan Dong
- Department of Cardiology, Shenzhen University General Hospital, Shenzhen, Guangdong, China
| | - Christof Stamm
- Berlin Institute of Health Center for Regenerative Therapies, Charité–Universitätsmedizin Berlin, Berlin, Germany
- Department of Cardiothoracic and Vascular Surgery, German Heart Center Berlin, Berlin, Germany
- German Centre for Cardiovascular Research, Partner Site Berlin, Berlin, Germany
- Helmholtz Zentrum Geesthacht, Institut für Aktive Polymere, Teltow, Germany
| |
Collapse
|
6
|
Evens L, Beliën H, D’Haese S, Haesen S, Verboven M, Rummens JL, Bronckaers A, Hendrikx M, Deluyker D, Bito V. Combinational Therapy of Cardiac Atrial Appendage Stem Cells and Pyridoxamine: The Road to Cardiac Repair? Int J Mol Sci 2021; 22:ijms22179266. [PMID: 34502175 PMCID: PMC8431115 DOI: 10.3390/ijms22179266] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/24/2021] [Accepted: 08/25/2021] [Indexed: 12/04/2022] Open
Abstract
Myocardial infarction (MI) occurs when the coronary blood supply is interrupted. As a consequence, cardiomyocytes are irreversibly damaged and lost. Unfortunately, current therapies for MI are unable to prevent progression towards heart failure. As the renewal rate of cardiomyocytes is minimal, the optimal treatment should achieve effective cardiac regeneration, possibly with stem cells transplantation. In that context, our research group identified the cardiac atrial appendage stem cells (CASCs) as a new cellular therapy. However, CASCs are transplanted into a hostile environment, with elevated levels of advanced glycation end products (AGEs), which may affect their regenerative potential. In this study, we hypothesize that pyridoxamine (PM), a vitamin B6 derivative, could further enhance the regenerative capacities of CASCs transplanted after MI by reducing AGEs’ formation. Methods and Results: MI was induced in rats by ligation of the left anterior descending artery. Animals were assigned to either no therapy (MI), CASCs transplantation (MI + CASCs), or CASCs transplantation supplemented with PM treatment (MI + CASCs + PM). Four weeks post-surgery, global cardiac function and infarct size were improved upon CASCs transplantation. Interstitial collagen deposition, evaluated on cryosections, was decreased in the MI animals transplanted with CASCs. Contractile properties of resident left ventricular cardiomyocytes were assessed by unloaded cell shortening. CASCs transplantation prevented cardiomyocyte shortening deterioration. Even if PM significantly reduced cardiac levels of AGEs, cardiac outcome was not further improved. Conclusion: Limiting AGEs’ formation with PM during an ischemic injury in vivo did not further enhance the improved cardiac phenotype obtained with CASCs transplantation. Whether AGEs play an important deleterious role in the setting of stem cell therapy after MI warrants further examination.
Collapse
Affiliation(s)
- Lize Evens
- UHasselt—Hasselt University, BIOMED—Biomedical Research Institute, Agoralaan, 3590 Diepenbeek, Belgium; (L.E.); (H.B.); (S.D.); (S.H.); (M.V.); (J.-L.R.); (A.B.); (M.H.); (D.D.)
| | - Hanne Beliën
- UHasselt—Hasselt University, BIOMED—Biomedical Research Institute, Agoralaan, 3590 Diepenbeek, Belgium; (L.E.); (H.B.); (S.D.); (S.H.); (M.V.); (J.-L.R.); (A.B.); (M.H.); (D.D.)
| | - Sarah D’Haese
- UHasselt—Hasselt University, BIOMED—Biomedical Research Institute, Agoralaan, 3590 Diepenbeek, Belgium; (L.E.); (H.B.); (S.D.); (S.H.); (M.V.); (J.-L.R.); (A.B.); (M.H.); (D.D.)
| | - Sibren Haesen
- UHasselt—Hasselt University, BIOMED—Biomedical Research Institute, Agoralaan, 3590 Diepenbeek, Belgium; (L.E.); (H.B.); (S.D.); (S.H.); (M.V.); (J.-L.R.); (A.B.); (M.H.); (D.D.)
| | - Maxim Verboven
- UHasselt—Hasselt University, BIOMED—Biomedical Research Institute, Agoralaan, 3590 Diepenbeek, Belgium; (L.E.); (H.B.); (S.D.); (S.H.); (M.V.); (J.-L.R.); (A.B.); (M.H.); (D.D.)
| | - Jean-Luc Rummens
- UHasselt—Hasselt University, BIOMED—Biomedical Research Institute, Agoralaan, 3590 Diepenbeek, Belgium; (L.E.); (H.B.); (S.D.); (S.H.); (M.V.); (J.-L.R.); (A.B.); (M.H.); (D.D.)
- UHasselt—Hasselt University, Faculty of Medicine and Life Sciences, Agoralaan, 3590 Diepenbeek, Belgium
| | - Annelies Bronckaers
- UHasselt—Hasselt University, BIOMED—Biomedical Research Institute, Agoralaan, 3590 Diepenbeek, Belgium; (L.E.); (H.B.); (S.D.); (S.H.); (M.V.); (J.-L.R.); (A.B.); (M.H.); (D.D.)
| | - Marc Hendrikx
- UHasselt—Hasselt University, BIOMED—Biomedical Research Institute, Agoralaan, 3590 Diepenbeek, Belgium; (L.E.); (H.B.); (S.D.); (S.H.); (M.V.); (J.-L.R.); (A.B.); (M.H.); (D.D.)
| | - Dorien Deluyker
- UHasselt—Hasselt University, BIOMED—Biomedical Research Institute, Agoralaan, 3590 Diepenbeek, Belgium; (L.E.); (H.B.); (S.D.); (S.H.); (M.V.); (J.-L.R.); (A.B.); (M.H.); (D.D.)
| | - Virginie Bito
- UHasselt—Hasselt University, BIOMED—Biomedical Research Institute, Agoralaan, 3590 Diepenbeek, Belgium; (L.E.); (H.B.); (S.D.); (S.H.); (M.V.); (J.-L.R.); (A.B.); (M.H.); (D.D.)
- Correspondence: ; Tel.: +32-11269285
| |
Collapse
|
7
|
Beliën H, Evens L, Hendrikx M, Bito V, Bronckaers A. Combining stem cells in myocardial infarction: The road to superior repair? Med Res Rev 2021; 42:343-373. [PMID: 34114238 DOI: 10.1002/med.21839] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 05/04/2021] [Accepted: 05/29/2021] [Indexed: 12/25/2022]
Abstract
Myocardial infarction irreversibly destroys millions of cardiomyocytes in the ventricle, making it the leading cause of heart failure worldwide. Over the past two decades, many progenitor and stem cell types were proposed as the ideal candidate to regenerate the heart after injury. The potential of stem cell therapy has been investigated thoroughly in animal and human studies, aiming at cardiac repair by true tissue replacement, by immune modulation, or by the secretion of paracrine factors that stimulate endogenous repair processes. Despite some successful results in animal models, the outcome from clinical trials remains overall disappointing, largely due to the limited stem cell survival and retention after transplantation. Extensive interest was developed regarding the combinational use of stem cells and various priming strategies to improve the efficacy of regenerative cell therapy. In this review, we provide a critical discussion of the different stem cell types investigated in preclinical and clinical studies in the field of cardiac repair. Moreover, we give an update on the potential of stem cell combinations as well as preconditioning and explore the future promises of these novel regenerative strategies.
Collapse
Affiliation(s)
- Hanne Beliën
- Biomedical Research Institute (BIOMED), Department of Cardio and Organ Systems, UHasselt-Hasselt University, Agoralaan, Diepenbeek, Belgium
| | - Lize Evens
- Biomedical Research Institute (BIOMED), Department of Cardio and Organ Systems, UHasselt-Hasselt University, Agoralaan, Diepenbeek, Belgium
| | - Marc Hendrikx
- Faculty of Medicine and Life Sciences, UHasselt-Hasselt University, Agoralaan, Diepenbeek, Belgium
| | - Virginie Bito
- Biomedical Research Institute (BIOMED), Department of Cardio and Organ Systems, UHasselt-Hasselt University, Agoralaan, Diepenbeek, Belgium
| | - Annelies Bronckaers
- Biomedical Research Institute (BIOMED), Department of Cardio and Organ Systems, UHasselt-Hasselt University, Agoralaan, Diepenbeek, Belgium
| |
Collapse
|
8
|
Elde S, Wang H, Woo YJ. Navigating the Crossroads of Cell Therapy and Natural Heart Regeneration. Front Cell Dev Biol 2021; 9:674180. [PMID: 34046410 PMCID: PMC8148343 DOI: 10.3389/fcell.2021.674180] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 04/15/2021] [Indexed: 01/14/2023] Open
Abstract
Cardiovascular disease remains the leading cause of death worldwide despite significant advances in our understanding of the disease and its treatment. Consequently, the therapeutic potential of cell therapy and induction of natural myocardial regeneration have stimulated a recent surge of research and clinical trials aimed at addressing this challenge. Recent developments in the field have shed new light on the intricate relationship between inflammation and natural regeneration, an intersection that warrants further investigation.
Collapse
Affiliation(s)
- Stefan Elde
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, United States
| | - Hanjay Wang
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, United States.,Stanford Cardiovascular Institute, Stanford University, Stanford, CA, United States
| | - Y Joseph Woo
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, United States.,Stanford Cardiovascular Institute, Stanford University, Stanford, CA, United States.,Department of Bioengineering, Stanford University, Stanford, CA, United States
| |
Collapse
|
9
|
Li J, Hu S, Zhu D, Huang K, Mei X, López de Juan Abad B, Cheng K. All Roads Lead to Rome (the Heart): Cell Retention and Outcomes From Various Delivery Routes of Cell Therapy Products to the Heart. J Am Heart Assoc 2021; 10:e020402. [PMID: 33821664 PMCID: PMC8174178 DOI: 10.1161/jaha.120.020402] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
In the past decades, numerous preclinical studies and several clinical trials have evidenced the feasibility of cell transplantation in treating heart diseases. Over the years, different delivery routes of cell therapy have emerged and broadened the width of the field. However, a common hurdle is shared by all current delivery routes: low cell retention. A myriad of studies confirm that cell retention plays a crucial role in the success of cell-mediated cardiac repair. It is important for any delivery route to maintain donor cells in the recipient heart for enough time to not only proliferate by themselves, but also to send paracrine signals to surrounding damaged heart cells and repair them. In this review, we first undertake an in-depth study of primary theories of cell loss, including low efficiency in cell injection, "washout" effects, and cell death, and then organize the literature from the past decade that focuses on cell transplantation to the heart using various cell delivery routes, including intracoronary injection, systemic intravenous injection, retrograde coronary venous injection, and intramyocardial injection. In addition to a recapitulation of these approaches, we also clearly evaluate their strengths and weaknesses. Furthermore, we conduct comparative research on the cell retention rate and functional outcomes of these delivery routes. Finally, we extend our discussion to state-of-the-art bioengineering techniques that enhance cell retention, as well as alternative delivery routes, such as intrapericardial delivery. A combination of these novel strategies and more accurate assessment methods will help to address the hurdle of low cell retention and boost the efficacy of cell transplantation to the heart.
Collapse
Affiliation(s)
- Junlang Li
- Department of Molecular Biomedical SciencesNorth Carolina State UniversityRaleighNC
- Joint Department of Biomedical EngineeringNorth Carolina State University and University of North Carolina at Chapel HillRaleighNC
| | - Shiqi Hu
- Department of Molecular Biomedical SciencesNorth Carolina State UniversityRaleighNC
- Joint Department of Biomedical EngineeringNorth Carolina State University and University of North Carolina at Chapel HillRaleighNC
| | - Dashuai Zhu
- Department of Molecular Biomedical SciencesNorth Carolina State UniversityRaleighNC
- Joint Department of Biomedical EngineeringNorth Carolina State University and University of North Carolina at Chapel HillRaleighNC
| | - Ke Huang
- Department of Molecular Biomedical SciencesNorth Carolina State UniversityRaleighNC
- Joint Department of Biomedical EngineeringNorth Carolina State University and University of North Carolina at Chapel HillRaleighNC
| | - Xuan Mei
- Department of Molecular Biomedical SciencesNorth Carolina State UniversityRaleighNC
- Joint Department of Biomedical EngineeringNorth Carolina State University and University of North Carolina at Chapel HillRaleighNC
| | - Blanca López de Juan Abad
- Department of Molecular Biomedical SciencesNorth Carolina State UniversityRaleighNC
- Joint Department of Biomedical EngineeringNorth Carolina State University and University of North Carolina at Chapel HillRaleighNC
| | - Ke Cheng
- Department of Molecular Biomedical SciencesNorth Carolina State UniversityRaleighNC
- Joint Department of Biomedical EngineeringNorth Carolina State University and University of North Carolina at Chapel HillRaleighNC
| |
Collapse
|
10
|
Lotfi F, Jafari M, Rezaei Hemami M, Salesi M, Nikfar S, Behnam Morshedi H, Kojuri J, Keshavarz K. Evaluation of the effectiveness of infusion of bone marrow derived cell in patients with heart failure: A network meta-analysis of randomized clinical trials and cohort studies. Med J Islam Repub Iran 2020; 34:178. [PMID: 33816377 PMCID: PMC8004572 DOI: 10.47176/mjiri.34.178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Indexed: 11/21/2022] Open
Abstract
Background: The aim of this study was to investigate the effectiveness of bone marrow-derived cells (BMC) technology in patients with heart failure and compare it with alternative therapies, including drug therapy, cardiac resynchronization therapy pacemaker (CRT-P), cardiac resynchronization therapy defibrillator (CRT-D).
Methods: A systematic review study was conducted to identify all clinical studies published by 2017. Using keywords such as "Heart Failure, BMC, Drug Therapy, CRT-D, CRT-P" and combinations of the mentioned words, we searched electronic databases, including Scopus, Cochrane Library, and PubMed. The quality of the selected studies was assessed using the Cochrane Collaboration's tool and the Newcastle-Ottawa. The primary and secondary end-points were left ventricular ejection fraction (LVEF) (%), failure cases (Number), left ventricular end-systolic volume (LVES) (ml), and left ventricular end-diastolic volume (LVED) (ml). Random-effects network meta-analyses were used to conduct a systematic comparison. Statistical analysis was done using STATA.
Results: This network meta-analysis covered a total of 57 final studies and 6694 patients. The Comparative effectiveness of BMC versus CRT-D, Drug, and CRT-P methods indicated the statistically significant superiority of BMC over CRT-P (6.607, 95% CI: 2.92, 10.29) in LVEF index and overall CRT-P (-13.946, 95% CI: -18.59, -9.29) and drug therapy (-4.176, 95% CI: -8.02, -.33) in LVES index. In addition, in terms of LVED index, the BMC had statistically significant differences with CRT-P (-10.187, 95% CI: -18.85, -1.52). BMC was also dominant to all methods in failure cases as a final outcome and the difference was statistically significant i.e. BMC vs CRT-D: 0.529 (0.45, 0.62) and BMC vs Drug: 0.516 (0.44, 0.60). In none of the outcomes, the other methods were statistically more efficacious than BMC. The BMC method was superior or similar to the other methods in all outcomes.
Conclusion: The results of this study showed that the BMC method, in general, and especially in terms of failure cases index, had a higher level of clinical effectiveness. However, due to the lack of data asymmetry, insufficient data and head-to-head studies, BMC in this meta-analysis might be considered as an alternative to existing treatments for heart failure.
Collapse
Affiliation(s)
- Farhad Lotfi
- Health Human Resources Research Center, School of Management and Medical Informatics, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mojtaba Jafari
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Mahmood Salesi
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Shekoufeh Nikfar
- Department of Pharmacoeconomics and Pharmaceutical Administration, Faculty of Pharmacy and Evidence-Based Medicine Group, Pharmaceutical Sciences Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Javad Kojuri
- Department of Cardiology, School of Medicine, Clinical Education Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Khosro Keshavarz
- Health Human Resources Research Center, School of Management and Medical Informatics, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
11
|
Weber F, Knapp G, Glass Ä, Kundt G, Ickstadt K. Interval estimation of the overall treatment effect in random-effects meta-analyses: Recommendations from a simulation study comparing frequentist, Bayesian, and bootstrap methods. Res Synth Methods 2020; 12:291-315. [PMID: 33264488 DOI: 10.1002/jrsm.1471] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 11/24/2020] [Accepted: 11/30/2020] [Indexed: 11/08/2022]
Abstract
There exists a variety of interval estimators for the overall treatment effect in a random-effects meta-analysis. A recent literature review summarizing existing methods suggested that in most situations, the Hartung-Knapp/Sidik-Jonkman (HKSJ) method was preferable. However, a quantitative comparison of those methods in a common simulation study is still lacking. Thus, we conduct such a simulation study for continuous and binary outcomes, focusing on the medical field for application. Based on the literature review and some new theoretical considerations, a practicable number of interval estimators is selected for this comparison: the classical normal-approximation interval using the DerSimonian-Laird heterogeneity estimator, the HKSJ interval using either the Paule-Mandel or the Sidik-Jonkman heterogeneity estimator, the Skovgaard higher-order profile likelihood interval, a parametric bootstrap interval, and a Bayesian interval using different priors. We evaluate the performance measures (coverage and interval length) at specific points in the parameter space, that is, not averaging over a prior distribution. In this sense, our study is conducted from a frequentist point of view. We confirm the main finding of the literature review, the general recommendation of the HKSJ method (here with the Sidik-Jonkman heterogeneity estimator). For meta-analyses including only two studies, the high length of the HKSJ interval limits its practical usage. In this case, the Bayesian interval using a weakly informative prior for the heterogeneity may help. Our recommendations are illustrated using a real-world meta-analysis dealing with the efficacy of an intramyocardial bone marrow stem cell transplantation during coronary artery bypass grafting.
Collapse
Affiliation(s)
- Frank Weber
- Institute for Biostatistics and Informatics in Medicine and Ageing Research, Rostock University Medical Center, Rostock, Germany
| | - Guido Knapp
- Department of Statistics, TU Dortmund University, Dortmund, Germany
| | - Änne Glass
- Institute for Biostatistics and Informatics in Medicine and Ageing Research, Rostock University Medical Center, Rostock, Germany
| | - Günther Kundt
- Institute for Biostatistics and Informatics in Medicine and Ageing Research, Rostock University Medical Center, Rostock, Germany
| | - Katja Ickstadt
- Department of Statistics, TU Dortmund University, Dortmund, Germany
| |
Collapse
|
12
|
Yamamoto S, Ding N, Matsumoto SI, Hirabayashi H. Highly specific, quantitative polymerase chain reaction probe for the quantification of human cells in cynomolgus monkeys. Drug Metab Pharmacokinet 2020; 36:100359. [PMID: 33348238 DOI: 10.1016/j.dmpk.2020.09.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 09/17/2020] [Accepted: 09/23/2020] [Indexed: 10/23/2022]
Abstract
Quantification of human cells may be performed using quantitative polymerase chain reaction (qPCR). In preclinical studies, the human Alu sequence is widely used as biomarker for human DNA. However, because the Alu gene is shared by primates, its use is limited to non-primate studies. The biodistribution of human cells in primates is also necessary for translational studies. Therefore, we aimed to design a novel, human-specific primer/probe that enables the quantification of human cells in primates and other animal models. A novel primer/probe set was successfully designed based on highly repetitive LINE1 sequences. qPCR efficiency (94.95-99.21%) and linearity of calibration curves (r2 = 0.996-0.999) were confirmed in tissue homogenates of cynomolgus monkey. The lower limit of detection was 10 cells per 15-mg tissue sample, a sensitivity that is equivalent to existing Alu primers/probes. The set was also effective in other animal models such as mice, rabbits, pigs, and common marmosets. To our knowledge, this is the first study describing the successful design of a human-specific qPCR primer/probe for human cell quantification in various animals, including non-human primates, using LINE1 sequence. The excellent selectivity, sensitivity, and versatility of the LINE1 primers/probes make it a promising quantification tool in preclinical biodistribution studies.
Collapse
Affiliation(s)
- Syunsuke Yamamoto
- Drug Metabolism and Pharmacokinetics Research Laboratories, Research, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa, Japan.
| | - Ning Ding
- Drug Metabolism and Pharmacokinetics Research Laboratories, Research, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa, Japan.
| | - Shin-Ichi Matsumoto
- Drug Metabolism and Pharmacokinetics Research Laboratories, Research, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa, Japan.
| | - Hideki Hirabayashi
- Drug Metabolism and Pharmacokinetics Research Laboratories, Research, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa, Japan.
| |
Collapse
|
13
|
Anttila V, Saraste A, Knuuti J, Jaakkola P, Hedman M, Svedlund S, Lagerström-Fermér M, Kjaer M, Jeppsson A, Gan LM. Synthetic mRNA Encoding VEGF-A in Patients Undergoing Coronary Artery Bypass Grafting: Design of a Phase 2a Clinical Trial. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2020; 18:464-472. [PMID: 32728595 PMCID: PMC7369517 DOI: 10.1016/j.omtm.2020.05.030] [Citation(s) in RCA: 93] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 05/27/2020] [Indexed: 12/16/2022]
Abstract
Therapeutic angiogenesis may improve outcomes in patients with coronary artery disease undergoing surgical revascularization. Angiogenic factors may promote blood vessel growth and regenerate regions of ischemic but viable myocardium. Previous clinical trials of vascular endothelial growth factor A (VEGF-A) gene therapy with DNA or viral vectors demonstrated safety but not efficacy. AZD8601 is VEGF-A165 mRNA formulated in biocompatible citrate-buffered saline and optimized for high-efficiency VEGF-A expression with minimal innate immune response. EPICCURE is an ongoing randomized, double-blind, placebo-controlled study of the safety of AZD8601 in patients with moderately decreased left ventricular function (ejection fraction 30%–50%) undergoing elective coronary artery bypass surgery. AZD8601 3 mg, 30 mg, or placebo is administered as 30 epicardial injections in a 10-min extension of cardioplegia. Injections are targeted to ischemic but viable myocardial regions in each patient using quantitative 15O-water positron emission tomography (PET) imaging (stress myocardial blood flow < 2.3 mL/g/min; resting myocardial blood flow > 0.6 mL/g/min). Improvement in regional and global myocardial blood flow quantified with 15O-water PET is an exploratory efficacy outcome, together with echocardiographic, clinical, functional, and biomarker measures. EPICCURE combines high-efficiency delivery with quantitative targeting and follow-up for robust assessment of the safety and exploratory efficacy of VEGF-A mRNA angiogenesis (ClinicalTrials.gov: NCT03370887).
Collapse
Affiliation(s)
- Vesa Anttila
- Heart Center, Turku University Hospital and University of Turku, Turku, Finland
| | - Antti Saraste
- Heart Center, Turku University Hospital and University of Turku, Turku, Finland.,Turku PET Center, Turku University Hospital and University of Turku, Turku, Finland
| | - Juhani Knuuti
- Turku PET Center, Turku University Hospital and University of Turku, Turku, Finland
| | - Pekka Jaakkola
- Heart Center, Kuopio University Hospital, Kuopio, Finland
| | - Marja Hedman
- Heart Center, Kuopio University Hospital, Kuopio, Finland
| | - Sara Svedlund
- Department of Clinical Physiology, Sahlgrenska University Hospital, Gothenburg, Sweden.,Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Maria Lagerström-Fermér
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Magnus Kjaer
- Early Biometrics and Statistical Innovation, Data Science & AI, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Anders Jeppsson
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Department of Cardiothoracic Surgery, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Li-Ming Gan
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden.,Department of Cardiology, Sahlgrenska University Hospital, Gothenburg, Sweden
| |
Collapse
|
14
|
How to Stimulate Myocardial Regeneration in Adult Mammalian Heart: Existing Views and New Approaches. BIOMED RESEARCH INTERNATIONAL 2020; 2020:7874109. [PMID: 32190680 PMCID: PMC7073483 DOI: 10.1155/2020/7874109] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Accepted: 02/13/2020] [Indexed: 12/19/2022]
Abstract
Stem cell-based therapy has been considered as a promising option in the treatment of ischemic heart disease. Although stem cell administration resulted in the temporary improvement of myocardial contractility in the majority of studies, the formation of new cardiomyocytes within the injured myocardium has not been conclusively demonstrated. Consequently, the focus of research in the field has since shifted to stem cell-derived paracrine factors, including cytokines, growth factors, mRNA, and miRNA. Notably, both mRNA and miRNA can enter into the extracellular space either in soluble form or packed into membrane vesicles. Stem cell-derived paracrine factors have been shown to suppress inflammation and apoptosis, stimulate angiogenesis, and amplify the proliferation and differentiation of resident cardiac stem cells (CSCs). Such features have led to exosomes being considered as potential drug candidates affording myocardial regeneration. The search for chemical signals capable of stimulating cardiomyogenesis is ongoing despite continuous debates regarding the ability of mature cardiomyocytes to divide or dedifferentiate, transdifferentiation of other cells into cardiomyocytes, and the ability of CSCs to differentiate into cardiomyocytes. Future research is aimed at identifying novel cell candidates capable of differentiating into cardiomyocytes. The observation that CSCs can undergo intracellular development with the formation of “cell-in-cell structure” and subsequent release of transitory amplifying cells with the capacity to differentiate into cardiomyocytes may provide clues for stimulating regenerative cardiomyogenesis.
Collapse
|
15
|
Abstract
The effects of cell therapy on heart regeneration in patients with chronic cardiomyopathy have been assessed in several clinical trials. These trials can be categorized as those using noncardiac stem cells, including mesenchymal stem cells, and those using cardiac-committed cells, including KIT+ cardiac stem cells, cardiosphere-derived cells, and cardiovascular progenitor cells derived from embryonic stem cells. Although the safety of cell therapies has been consistently reported, their efficacy remains more elusive. Nevertheless, several lessons have been learned that provide useful clues for future studies. This Review summarizes the main outcomes of these studies and draws some perspectives for future cell-based regenerative trials, which are largely based on the primary therapeutic target: remuscularization of chronic myocardial scars by exogenous cells or predominant use of these cells to activate host-associated repair pathways though paracrine signalling. In the first case, the study design should entail delivery of large numbers of cardiac-committed cells, supply of supportive noncardiac cells, and promotion of cell survival and appropriate coupling with endogenous cardiomyocytes. If the primary objective is to harness endogenous repair pathways, then the flexibility of cell type is greater. As the premise is that the transplanted cells need to engraft only transiently, the priority is to optimize their early retention and possibly to switch towards the sole administration of their secretome.
Collapse
|
16
|
Pooria A, Pourya A, Gheini A. Animal- and human-based evidence for the protective effects of stem cell therapy against cardiovascular disorders. J Cell Physiol 2019; 234:14927-14940. [PMID: 30811030 DOI: 10.1002/jcp.28330] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 12/06/2018] [Accepted: 01/22/2019] [Indexed: 01/24/2023]
Abstract
The increasing rate of mortality and morbidity because of cardiac diseases has called for efficient therapeutic needs. With the advancement in cell-based therapies, stem cells are abundantly studied in this area. Nearly, all sources of stem cells are experimented to treat cardiac injuries. Tissue engineering has also backed this technique by providing an advantageous platform to improve stem cell therapy. After in vitro studies, primary treatment-based research studies comprise small and large animal studies. Furthermore, these studies are implemented in human models in the form of clinical trials. Purpose of this review is to highlight the animal- and human-based studies, exploiting various stem cell sources, to treat cardiovascular disorders.
Collapse
Affiliation(s)
- Ali Pooria
- Department of Cardiology, Lorestan University of Medical Sciences, Khoramabad, Iran
| | - Afsoun Pourya
- Student of Research committee, Tehran University of Medical Sciences, Tehran, Iran
| | - Alireza Gheini
- Department of Cardiology, Lorestan University of Medical Sciences, Khoramabad, Iran
| |
Collapse
|
17
|
Combined Coronary Artery Bypass Surgery With Bone Marrow Stem Cell Transplantation: Are We There Yet? Ann Thorac Surg 2019; 108:1913-1921. [PMID: 31398356 DOI: 10.1016/j.athoracsur.2019.05.094] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Revised: 04/11/2019] [Accepted: 05/20/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND Although the safety and feasibility of combined coronary artery bypass grafting (CABG) and bone marrow stem cell (BMSC) transplantation have been proven, the efficacy of this approach remains controversial. Therefore, we conducted an updated meta-analysis of randomized controlled trials to evaluate the efficacy of this procedure. METHODS Electronic databases were systematically searched for randomized trials comparing 4-month to 6-month follow-up outcomes in patients who underwent isolated CABG (CABG group) and patients who received BMSC transplantation with CABG (BMSC group). A random-effects meta-analysis was conducted across eligible studies. Meta-regression and subgroup analyses were utilized to identify sources of data heterogeneity. RESULTS Thirteen trials were eligible, with a total number of 292 patients in the BMSC group and 247 patients in the CABG group. Compared with the CABG group, the BMSC group showed significant improvement of follow-up left ventricular ejection fraction (n = 539, 4.8%; 95% confidence interval [CI], 2.3%-7.3%; P = .001). The analyzed data showed significant heterogeneity (I2 = 74.2%, P < .001). The reduction in scar size (n = 120; -2.2 mL; 95% CI, -18.2 mL to 13.7 mL; P = .44) and the improvement in the 6-minute walk test (n = 212; 41 m; 95% CI, -13 m to 95 m; P = .10) did not reach statistical significance. No significant correlation was found between the number of the injected BMSCs or the method of injection and the change in ejection fraction. CONCLUSIONS The present evidence suggests that combined CABG and BMSC transplantation is associated with improvement of left ventricular ejection fraction. However, the heterogeneity in the data suggests variations in patient response to this therapy. Further studies are required to understand these variations.
Collapse
|
18
|
Wu S, Yao L, Yan P, Bao Q, Dong X, Liu X, Zhu Y, Su X, Wang A, Duan Y, Yang K, Zhang M, Cao Y. Autologous bone marrow stem cell therapy for patients undergoing coronary artery bypass grafting: A meta-analysis of 14 randomized controlled trials. Exp Ther Med 2019; 17:2985-2994. [PMID: 30906476 PMCID: PMC6425237 DOI: 10.3892/etm.2019.7283] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 01/24/2019] [Indexed: 01/01/2023] Open
Abstract
Autologous bone marrow stem cell (BMSC) therapy is a novel option for regenerative therapy in patients with ischemic heart disease. The aim of the present meta-analysis was to evaluate the effectiveness of BMSCs combined with coronary artery bypass grafting (CABG). The PubMed, Cochrane Library, EMBASE and Web of Science databases were searched from inception to November 22, 2017 for randomized controlled trials on BMSC therapy combined with CABG. Finally, 14 trials with a total of 596 participants were included. Data were analyzed using a random-effects model. Compared with the control group, the BMSC therapy group exhibited an improvement in the left ventricular (LV) ejection fraction from baseline to follow-up [mean difference (MD)=4.36%; 95% confidence interval (CI): 1.90–6.81%; P<0.01]. Analysis of the pooled results revealed non-significant differences in the LV end-diastolic volume (MD=−6.27 ml; 95% CI: −22.34 to 9.80 ml; P=0.44), LV end-diastolic volume index (MD=−15.11 ml/m2; 95% CI: −31.53 to 1.30 ml/m2; P=0.07), LV end-systolic volume (MD=−11.52 ml; 95% CI: −26.97 to 3.93 ml; P=0.14) and LV end-systolic volume index (MD=−16.56 ml/m2; 95% CI: −37.75 to 4.63 ml/m2; P=0.13) between the BMSC and CABG alone groups. Therefore, autologous BMSC therapy for patients undergoing CABG appears to be associated with an improvement in LV function compared with CABG alone.
Collapse
Affiliation(s)
- Shanlian Wu
- Department of Pathology, School of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu 730000, P.R. China.,Department of Pathology, Gansu Provincial Hospital, Lanzhou, Gansu 730000, P.R. China.,Institute of Clinical Research and Evidence-Based Medicine, Gansu Provincial Hospital, Lanzhou, Gansu 730000, P.R. China.,Department of Evidence-Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Liang Yao
- Institute of Clinical Research and Evidence-Based Medicine, Gansu Provincial Hospital, Lanzhou, Gansu 730000, P.R. China
| | - Peijing Yan
- Institute of Clinical Research and Evidence-Based Medicine, Gansu Provincial Hospital, Lanzhou, Gansu 730000, P.R. China
| | - Qiangwei Bao
- Department of Pathology, School of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu 730000, P.R. China.,Department of Pathology, Gansu Provincial Hospital, Lanzhou, Gansu 730000, P.R. China
| | - Xin Dong
- Department of Ultrasound, The Third Hospital of Gansu Province, Lanzhou, Gansu 730020, P.R. China
| | - Xingguang Liu
- Department of Cardiac Surgery, Gansu Provincial Hospital, Lanzhou, Gansu 730000, P.R. China
| | - Yan Zhu
- Department of Cardiology, Gansu Provincial Hospital, Lanzhou, Gansu 730000, P.R. China
| | - Xin Su
- Department of Cardiology, Gansu Provincial Hospital, Lanzhou, Gansu 730000, P.R. China.,Department of Cardiology, School of Clinical Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu 730000, P.R. China
| | - Aqian Wang
- Department of Cardiology, Gansu Provincial Hospital, Lanzhou, Gansu 730000, P.R. China.,Department of Cardiology, School of Clinical Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu 730000, P.R. China
| | - Yichao Duan
- Department of Cardiology, Gansu Provincial Hospital, Lanzhou, Gansu 730000, P.R. China
| | - Kehu Yang
- Institute of Clinical Research and Evidence-Based Medicine, Gansu Provincial Hospital, Lanzhou, Gansu 730000, P.R. China.,Department of Evidence-Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Min Zhang
- Department of Pathology, Gansu Provincial Hospital, Lanzhou, Gansu 730000, P.R. China
| | - Yunshan Cao
- Department of Cardiology, Gansu Provincial Hospital, Lanzhou, Gansu 730000, P.R. China
| |
Collapse
|
19
|
Mann I, Tseng CCS, Rodrigo SF, Koudstaal S, van Ramshorst J, Beeres SL, Dibbets-Schneider P, de Geus-Oei LF, Lamb HJ, Wolterbeek R, Zwaginga JJ, Fibbe WE, Westinga K, Bax JJ, Doevendans PA, Schalij MJ, Chamuleau SAJ, Atsma DE. Intramyocardial bone marrow cell injection does not lead to functional improvement in patients with chronic ischaemic heart failure without considerable ischaemia. Neth Heart J 2018; 27:81-92. [PMID: 30569306 PMCID: PMC6352621 DOI: 10.1007/s12471-018-1213-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Background It has been suggested that bone marrow cell injection may have beneficial effects in patients with chronic ischaemic heart disease. However, previous trials have led to discrepant results of cell-based therapy in patients with chronic heart failure. The aim of this study was to evaluate the efficacy of intramyocardial injection of mononuclear bone marrow cells in patients with chronic ischaemic heart failure with limited stress-inducible myocardial ischaemia. Methods and results This multicentre, randomised, placebo-controlled trial included 39 patients with no-option chronic ischaemic heart failure with a follow-up of 12 months. A total of 19 patients were randomised to autologous intramyocardial bone marrow cell injection (cell group) and 20 patients received a placebo injection (placebo group). The primary endpoint was the group difference in change of left ventricular ejection fraction, as determined by single-photon emission tomography. On follow-up at 3 and 12 months, change of left ventricular ejection fraction in the cell group was comparable with change in the placebo group (P = 0.47 and P = 0.08, respectively). Also secondary endpoints, including left ventricle volumes, myocardial perfusion, functional and clinical parameters did not significantly change in the cell group as compared to placebo. Neither improvement was demonstrated in a subgroup of patients with stress-inducible ischaemia (P = 0.54 at 3‑month and P = 0.15 at 12-month follow-up). Conclusion Intramyocardial bone marrow cell injection does not improve cardiac function, nor functional and clinical parameters in patients with severe chronic ischaemic heart failure with limited stress-inducible ischaemia. Clinical Trial Registration: NTR2516 Electronic supplementary material The online version of this article (10.1007/s12471-018-1213-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- I Mann
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| | - C C S Tseng
- Department of Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - S F Rodrigo
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| | - S Koudstaal
- Department of Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - J van Ramshorst
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| | - S L Beeres
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| | - P Dibbets-Schneider
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - L F de Geus-Oei
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands.,MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands
| | - H J Lamb
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - R Wolterbeek
- Department of Medical Statistics and Bioinformatics, Leiden University Medical Center, Leiden, The Netherlands
| | - J J Zwaginga
- Department of Hematology, Leiden University Medical Center, Leiden, The Netherlands
| | - W E Fibbe
- Department of Hematology, Leiden University Medical Center, Leiden, The Netherlands
| | - K Westinga
- Department of Cell Therapy Facility, University Medical Center Utrecht, Utrecht, The Netherlands
| | - J J Bax
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| | - P A Doevendans
- Department of Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - M J Schalij
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| | - S A J Chamuleau
- Department of Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - D E Atsma
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands.
| |
Collapse
|
20
|
Michler RE. The role of stem cells in treating coronary artery disease in 2018. Indian J Thorac Cardiovasc Surg 2018; 34:340-348. [PMID: 33060957 DOI: 10.1007/s12055-018-0739-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 08/24/2018] [Accepted: 08/27/2018] [Indexed: 11/27/2022] Open
Abstract
The last decade has witnessed the publication of a number of stem cell clinical trials, primarily using bone marrow-derived cells as the injected cell. Much has been learned through these "first-generation" clinical trials. The advances in our understanding include the following: (1) cell therapy is safe; (2) cell therapy has been mildly effective; and (3) human bone marrow-derived stem cells do not transdifferentiate into cardiomyocytes or new blood vessels. The primary mechanism of action for cell therapy is now believed to be through paracrine effects that include the release of cytokines, chemokines, and growth factors that inhibit apoptosis and fibrosis, enhance contractility, and activate endogenous regenerative mechanisms through endogenous circulating or site-specific stem cells. The current direction for clinical trials includes the use of stem cells capable of cardiac lineage.
Collapse
Affiliation(s)
- Robert E Michler
- Department of Surgery, Montefiore Medical Center, Albert Einstein College of Medicine, Greene Medical Arts Pavilion 5th Floor, 3400 Bainbridge Avenue, New York City, NY 10467 USA
- Department of Cardiothoracic & Vascular Surgery, Montefiore Medical Center, Albert Einstein College of Medicine, Greene Medical Arts Pavilion 5th Floor, 3400 Bainbridge Avenue, New York City, NY 10467 USA
| |
Collapse
|
21
|
Michler RE. The current status of stem cell therapy in ischemic heart disease. J Card Surg 2018; 33:520-531. [DOI: 10.1111/jocs.13789] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Robert E. Michler
- Department of Cardiothoracic and Vascular Surgery and Department of Surgery; Montefiore Medical Center, Albert Einstein College of Medicine; New York New York
| |
Collapse
|
22
|
Tanaka R, Masuda H, Fujimura S, Ito-Hirano R, Arita K, Kakinuma Y, Hagiwara H, Kado M, Hayashi A, Mita T, Ogawa T, Watada H, Mizuno H, Sawada N, Asahara T. Quality-Quantity Control Culture Enhances Vasculogenesis and Wound Healing Efficacy of Human Diabetic Peripheral Blood CD34+ Cells. Stem Cells Transl Med 2018; 7:428-438. [PMID: 29573563 PMCID: PMC5905232 DOI: 10.1002/sctm.17-0043] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2017] [Accepted: 01/17/2018] [Indexed: 01/16/2023] Open
Abstract
Autologous endothelial progenitor cell (EPC) therapy is commonly used to stimulate angiogenesis in ischemic repair and wound healing. However, low total numbers and functional deficits of EPCs make autologous EPC therapy ineffective in diabetes. Currently, no known ex vivo culture techniques can expand and/or ameliorate the functional deficits of EPCs for clinical usage. Recently, we showed that a quality‐quantity culture (QQc) system restores the vasculogenic and wound‐healing efficacy of murine diabetic EPCs. To validate these results and elucidate the mechanism in a translational study, we evaluated the efficacy of this QQc system to restore the vasculogenic potential of diabetic human peripheral blood (PB) CD34+ cells. CD34+ cells purified from PB of diabetic and healthy patients were subjected to QQc. Gene expression, vascular regeneration, and expression of cytokines and paracrine mediators were analyzed. Pre‐ or post‐QQc diabetic human PB‐CD34+ cells were transplanted into wounded BALB/c nude mice and streptozotocin‐induced diabetic mice to assess functional efficacy. Post‐QQc diabetic human PB‐CD34+ cell therapy significantly accelerated wound closure, re‐epithelialization, and angiogenesis. The higher therapeutic efficacy of post‐QQc diabetic human PB‐CD34+ cells was attributed to increased differentiation ability of diabetic CD34+ cells, direct vasculogenesis, and enhanced expression of angiogenic factors and wound‐healing genes. Thus, QQc can significantly enhance the therapeutic efficacy of human PB‐CD34+ cells in diabetic wounds, overcoming the inherent limitation of autologous cell therapy in diabetic patients, and could be useful for treatment of not only wounds but also other ischemic diseases. Stem Cells Translational Medicine2018;7:428–438
Collapse
Affiliation(s)
- Rica Tanaka
- Department of Plastic and Reconstructive Surgery, Tokyo, Japan
| | - Haruchika Masuda
- Department of Basic Clinical Science, Division of Regenerative Medicine, Tokai University School of Medicine, Isehara, Japan
| | | | - Rie Ito-Hirano
- Department of Plastic and Reconstructive Surgery, Tokyo, Japan
| | - Kayo Arita
- Department of Plastic and Reconstructive Surgery, Tokyo, Japan
| | - Yusuke Kakinuma
- Department of Plastic and Reconstructive Surgery, Tokyo, Japan
| | - Hiroko Hagiwara
- Department of Plastic and Reconstructive Surgery, Tokyo, Japan
| | - Makiko Kado
- Department of Plastic and Reconstructive Surgery, Tokyo, Japan
| | - Ayato Hayashi
- Department of Plastic and Reconstructive Surgery, Tokyo, Japan
| | - Tomoya Mita
- Department of Internal Medicine, Division of Diabetes and Metabolism, Tokyo, Japan
| | - Takasuke Ogawa
- Department of Dermatology, Juntendo University School of Medicine, Tokyo, Japan
| | - Hirotaka Watada
- Department of Internal Medicine, Division of Diabetes and Metabolism, Tokyo, Japan
| | - Hiroshi Mizuno
- Department of Plastic and Reconstructive Surgery, Tokyo, Japan
| | - Naoki Sawada
- Global COE Program, Tokyo Medical and Dental University, Tokyo, Japan
| | - Takayuki Asahara
- Department of Basic Clinical Science, Division of Regenerative Medicine, Tokai University School of Medicine, Isehara, Japan
| |
Collapse
|
23
|
Tang JN, Cores J, Huang K, Cui XL, Luo L, Zhang JY, Li TS, Qian L, Cheng K. Concise Review: Is Cardiac Cell Therapy Dead? Embarrassing Trial Outcomes and New Directions for the Future. Stem Cells Transl Med 2018; 7:354-359. [PMID: 29468830 PMCID: PMC5866934 DOI: 10.1002/sctm.17-0196] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 01/03/2018] [Accepted: 01/09/2018] [Indexed: 01/08/2023] Open
Abstract
Stem cell therapy is a promising strategy for tissue regeneration. The therapeutic benefits of cell therapy are mediated by both direct and indirect mechanisms. However, the application of stem cell therapy in the clinic is hampered by several limitations. This concise review provides a brief introduction into stem cell therapies for ischemic heart disease. It summarizes cell‐based and cell‐free paradigms, their limitations, and the benefits of using them to target disease. stemcellstranslationalmedicine2018;7:354–359
Collapse
Affiliation(s)
- Jun-Nan Tang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China.,Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina, USA.,Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, North Carolina, USA
| | - Jhon Cores
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina, USA.,Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, North Carolina, USA
| | - Ke Huang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China.,Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina, USA
| | - Xiao-Lin Cui
- Department of Orthopaedic Surgery, University of Otago, Christchurch, New Zealand
| | - Lan Luo
- Department of Stem Cell Biology, Atomic Bomb Disease Institute, Nagasaki University, Nagasaki, Japan
| | - Jin-Ying Zhang
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, North Carolina, USA
| | - Tao-Sheng Li
- Department of Stem Cell Biology, Atomic Bomb Disease Institute, Nagasaki University, Nagasaki, Japan
| | - Li Qian
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Ke Cheng
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China.,Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina, USA.,Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, North Carolina, USA.,Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
24
|
Fernández-Avilés F, Sanz-Ruiz R, Climent AM, Badimon L, Bolli R, Charron D, Fuster V, Janssens S, Kastrup J, Kim HS, Lüscher TF, Martin JF, Menasché P, Simari RD, Stone GW, Terzic A, Willerson JT, Wu JC. Global position paper on cardiovascular regenerative medicine. Eur Heart J 2017; 38:2532-2546. [PMID: 28575280 PMCID: PMC5837698 DOI: 10.1093/eurheartj/ehx248] [Citation(s) in RCA: 115] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 04/13/2017] [Accepted: 04/20/2017] [Indexed: 12/11/2022] Open
Affiliation(s)
- Francisco Fernández-Avilés
- Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, Universidad Complutense, Madrid, Spain
- CIBERCV, ISCIII, Madrid, Spain
| | - Ricardo Sanz-Ruiz
- Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, Universidad Complutense, Madrid, Spain
- CIBERCV, ISCIII, Madrid, Spain
| | - Andreu M Climent
- Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, Universidad Complutense, Madrid, Spain
- CIBERCV, ISCIII, Madrid, Spain
| | - Lina Badimon
- CIBERCV, ISCIII, Madrid, Spain
- Cardiovascular Research Center (CSIC-ICCC), Hospital de la Santa Creu i Sant Pau (HSCSP), Barcelona, Spain
| | - Roberto Bolli
- Institute of Molecular Cardiology, Diabetes and Obesity Center, University of Louisville School of Medicine, Louisville, Kentucky
| | - Dominique Charron
- LabEx TRANSPLANTEX; HLA & Médecine "Jean Dausset" Laboratory Network, Hôpital Saint-Louis AP-HP, Université Paris Diderot, 75013, France
| | - Valentin Fuster
- CIBERCV, ISCIII, Madrid, Spain
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
- Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of medicine at Mount Sinai, New York, NY, USA
| | - Stefan Janssens
- Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - Jens Kastrup
- Department of Cardiology, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Hyo-Soo Kim
- National Research Laboratory for Stem Cell Niche, Center for Medical Innovation, Seoul National University Hospital, Seoul, Korea; Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Korea
| | - Thomas F Lüscher
- Department of Cardiology, University Heart Center Zurich, Zurich, Switzerland; Center for Molecular Cardiology, University of Zurich, Zurich, Switzerland
| | | | - Philippe Menasché
- Department of Cardiovascular Surgery Hôpital Européen Georges Pompidou; Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Robert D Simari
- School of Medicine, University of Kansas, 3901 Rainbow Boulevard, Kansas City, KS, USA
| | - Gregg W Stone
- Center for Clinical Trials, Cardiovascular Research Foundation, New York, New York; Center for Clinical Trials, NewYork-Presbyterian Hospital, Columbia University Medical Center, New York, NY, USA
| | - Andre Terzic
- Center for Regenerative Medicine, Department of Cardiovascular Diseases, Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, NY, USA
| | - James T Willerson
- Department of Regenerative Medicine Research, Texas Heart Institute, Houston, TX, USA
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Division of Cardiovascular Medicine, Department of Medicine and Department of Radiology, Stanford University School of Medicine, CA, USA
| |
Collapse
|
25
|
Cambria E, Pasqualini FS, Wolint P, Günter J, Steiger J, Bopp A, Hoerstrup SP, Emmert MY. Translational cardiac stem cell therapy: advancing from first-generation to next-generation cell types. NPJ Regen Med 2017; 2:17. [PMID: 29302353 PMCID: PMC5677990 DOI: 10.1038/s41536-017-0024-1] [Citation(s) in RCA: 93] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2016] [Revised: 05/16/2017] [Accepted: 05/22/2017] [Indexed: 12/16/2022] Open
Abstract
Acute myocardial infarction and chronic heart failure rank among the major causes of morbidity and mortality worldwide. Except for heart transplantation, current therapy options only treat the symptoms but do not cure the disease. Stem cell-based therapies represent a possible paradigm shift for cardiac repair. However, most of the first-generation approaches displayed heterogeneous clinical outcomes regarding efficacy. Stemming from the desire to closely match the target organ, second-generation cell types were introduced and rapidly moved from bench to bedside. Unfortunately, debates remain around the benefit of stem cell therapy, optimal trial design parameters, and the ideal cell type. Aiming at highlighting controversies, this article provides a critical overview of the translation of first-generation and second-generation cell types. It further emphasizes the importance of understanding the mechanisms of cardiac repair and the lessons learned from first-generation trials, in order to improve cell-based therapies and to potentially finally implement cell-free therapies.
Collapse
Affiliation(s)
- Elena Cambria
- Institute for Regenerative Medicine, University of Zurich, Zurich, 8044 Switzerland.,Division of Surgical Research, University Hospital of Zurich, Zurich, 8091 Switzerland
| | | | - Petra Wolint
- Institute for Regenerative Medicine, University of Zurich, Zurich, 8044 Switzerland.,Division of Surgical Research, University Hospital of Zurich, Zurich, 8091 Switzerland
| | - Julia Günter
- Institute for Regenerative Medicine, University of Zurich, Zurich, 8044 Switzerland.,Division of Surgical Research, University Hospital of Zurich, Zurich, 8091 Switzerland
| | - Julia Steiger
- Institute for Regenerative Medicine, University of Zurich, Zurich, 8044 Switzerland.,Division of Surgical Research, University Hospital of Zurich, Zurich, 8091 Switzerland
| | - Annina Bopp
- Institute for Regenerative Medicine, University of Zurich, Zurich, 8044 Switzerland.,Division of Surgical Research, University Hospital of Zurich, Zurich, 8091 Switzerland
| | - Simon P Hoerstrup
- Institute for Regenerative Medicine, University of Zurich, Zurich, 8044 Switzerland.,Division of Surgical Research, University Hospital of Zurich, Zurich, 8091 Switzerland.,Heart Center Zurich, University Hospital of Zurich, Zurich, Switzerland.,Wyss Translational Center Zurich, Zurich, Switzerland
| | - Maximilian Y Emmert
- Institute for Regenerative Medicine, University of Zurich, Zurich, 8044 Switzerland.,Division of Surgical Research, University Hospital of Zurich, Zurich, 8091 Switzerland.,Heart Center Zurich, University Hospital of Zurich, Zurich, Switzerland.,Wyss Translational Center Zurich, Zurich, Switzerland
| |
Collapse
|
26
|
Chen HY, Strappe PM, Wang LX. Stem Cell Therapies for Cardiovascular Diseases: What Does the Future Hold? Heart Lung Circ 2017; 26:205-208. [DOI: 10.1016/j.hlc.2016.12.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
27
|
Nigro P, Bassetti B, Cavallotti L, Catto V, Carbucicchio C, Pompilio G. Cell therapy for heart disease after 15 years: Unmet expectations. Pharmacol Res 2017; 127:77-91. [PMID: 28235633 DOI: 10.1016/j.phrs.2017.02.015] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 02/07/2017] [Accepted: 02/16/2017] [Indexed: 12/17/2022]
Abstract
Over the past two decades cardiac cell therapy (CCT) has emerged as a promising new strategy to cure heart diseases at high unmet need. Thousands of patients have entered clinical trials for acute or chronic heart conditions testing different cell types, including autologous or allogeneic bone marrow (BM)-derived mononuclear or selected cells, BM- or adipose tissue-derived mesenchymal cells, or cardiac resident progenitors based on their potential ability to regenerate scarred or dysfunctional myocardium. Nowadays, the original enthusiasm surrounding the regenerative medicine field has been cushioned by a cumulative body of evidence indicating an inefficient or modest efficacy of CCT in improving cardiac function, along with the continued lack of indisputable proof for long-term prognostic benefit. In this review, we have firstly comprehensively outlined the positive and negative results of cell therapy studies in patients with acute myocardial infarction, refractory angina and chronic heart failure. Next, we have discussed cell therapy- and patient-related variables (e.g. cell intrinsic and extrinsic characteristics as well as criteria of patient selection and proposed methodologies) that might have dampened the efficacy of past cell therapy trials. Finally, we have addressed critical factors to be considered before embarking on further clinical trials.
Collapse
Affiliation(s)
- Patrizia Nigro
- Vascular Biology and Regenerative Medicine Unit, Centro Cardiologico Monzino-IRCCS, via Carlo Parea 4, 20138, Milan, Italy
| | - Beatrice Bassetti
- Vascular Biology and Regenerative Medicine Unit, Centro Cardiologico Monzino-IRCCS, via Carlo Parea 4, 20138, Milan, Italy
| | - Laura Cavallotti
- Department of Cardiovascular Surgery, Centro Cardiologico Monzino-IRCCS, via Carlo Parea 4, 20138, Milan, Italy
| | - Valentina Catto
- Cardiac Arrhythmia Research Centre, Centro Cardiologico Monzino-IRCCS, via Carlo Parea 4, 20138, Milan, Italy
| | - Corrado Carbucicchio
- Cardiac Arrhythmia Research Centre, Centro Cardiologico Monzino-IRCCS, via Carlo Parea 4, 20138, Milan, Italy
| | - Giulio Pompilio
- Vascular Biology and Regenerative Medicine Unit, Centro Cardiologico Monzino-IRCCS, via Carlo Parea 4, 20138, Milan, Italy; Department of Clinical Sciences and Community Health, University of Milan, via Festa del Perdono 7, 20122, Milan, Italy.
| |
Collapse
|
28
|
Bone Marrow Cell Therapy for Ischemic Heart Disease and the Role of Cardiac Imaging in Evaluation of Outcomes. STEM CELL BIOLOGY AND REGENERATIVE MEDICINE 2017. [DOI: 10.1007/978-3-319-51833-6_9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
29
|
Fisher SA, Doree C, Mathur A, Taggart DP, Martin‐Rendon E. Stem cell therapy for chronic ischaemic heart disease and congestive heart failure. Cochrane Database Syst Rev 2016; 12:CD007888. [PMID: 28012165 PMCID: PMC6463978 DOI: 10.1002/14651858.cd007888.pub3] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
BACKGROUND A promising approach to the treatment of chronic ischaemic heart disease and congestive heart failure is the use of stem cells. The last decade has seen a plethora of randomised controlled trials developed worldwide, which have generated conflicting results. OBJECTIVES The critical evaluation of clinical evidence on the safety and efficacy of autologous adult bone marrow-derived stem/progenitor cells as a treatment for chronic ischaemic heart disease and congestive heart failure. SEARCH METHODS We searched CENTRAL in the Cochrane Library, MEDLINE, Embase, CINAHL, LILACS, and four ongoing trial databases for relevant trials up to 14 December 2015. SELECTION CRITERIA Eligible studies were randomised controlled trials comparing autologous adult stem/progenitor cells with no cells in people with chronic ischaemic heart disease and congestive heart failure. We included co-interventions, such as primary angioplasty, surgery, or administration of stem cell mobilising agents, when administered to treatment and control arms equally. DATA COLLECTION AND ANALYSIS Two review authors independently screened all references for eligibility, assessed trial quality, and extracted data. We undertook a quantitative evaluation of data using random-effects meta-analyses. We evaluated heterogeneity using the I2 statistic and explored substantial heterogeneity (I2 greater than 50%) through subgroup analyses. We assessed the quality of the evidence using the GRADE approach. We created a 'Summary of findings' table using GRADEprofiler (GRADEpro), excluding studies with a high or unclear risk of selection bias. We focused our summary of findings on long-term follow-up of mortality, morbidity outcomes, and left ventricular ejection fraction measured by magnetic resonance imaging. MAIN RESULTS We included 38 randomised controlled trials involving 1907 participants (1114 cell therapy, 793 controls) in this review update. Twenty-three trials were at high or unclear risk of selection bias. Other sources of potential bias included lack of blinding of participants (12 trials) and full or partial commercial sponsorship (13 trials).Cell therapy reduced the incidence of long-term mortality (≥ 12 months) (risk ratio (RR) 0.42, 95% confidence interval (CI) 0.21 to 0.87; participants = 491; studies = 9; I2 = 0%; low-quality evidence). Periprocedural adverse events associated with the mapping or cell/placebo injection procedure were infrequent. Cell therapy was also associated with a long-term reduction in the incidence of non-fatal myocardial infarction (RR 0.38, 95% CI 0.15 to 0.97; participants = 345; studies = 5; I2 = 0%; low-quality evidence) and incidence of arrhythmias (RR 0.42, 95% CI 0.18 to 0.99; participants = 82; studies = 1; low-quality evidence). However, we found no evidence that cell therapy affects the risk of rehospitalisation for heart failure (RR 0.63, 95% CI 0.36 to 1.09; participants = 375; studies = 6; I2 = 0%; low-quality evidence) or composite incidence of mortality, non-fatal myocardial infarction, and/or rehospitalisation for heart failure (RR 0.64, 95% CI 0.38 to 1.08; participants = 141; studies = 3; I2 = 0%; low-quality evidence), or long-term left ventricular ejection fraction when measured by magnetic resonance imaging (mean difference -1.60, 95% CI -8.70 to 5.50; participants = 25; studies = 1; low-quality evidence). AUTHORS' CONCLUSIONS This systematic review and meta-analysis found low-quality evidence that treatment with bone marrow-derived stem/progenitor cells reduces mortality and improves left ventricular ejection fraction over short- and long-term follow-up and may reduce the incidence of non-fatal myocardial infarction and improve New York Heart Association (NYHA) Functional Classification in people with chronic ischaemic heart disease and congestive heart failure. These findings should be interpreted with caution, as event rates were generally low, leading to a lack of precision.
Collapse
Affiliation(s)
- Sheila A Fisher
- NHS Blood and TransplantSystematic Review InitiativeLevel 2, John Radcliffe HospitalHeadingtonOxfordOxonUKOX3 9BQ
| | - Carolyn Doree
- NHS Blood and TransplantSystematic Review InitiativeLevel 2, John Radcliffe HospitalHeadingtonOxfordOxonUKOX3 9BQ
| | - Anthony Mathur
- William Harvey Research InstituteDepartment of Clinical PharmacologyCharterhouse SquareLondonUKEC1M 6BQ
| | | | - Enca Martin‐Rendon
- Radcliffe Department of Medicine, University of OxfordSystematic Review InitiativeOxfordUK
| | | |
Collapse
|
30
|
Psaltis PJ, Schwarz N, Toledo-Flores D, Nicholls SJ. Cellular Therapy for Heart Failure. Curr Cardiol Rev 2016; 12:195-215. [PMID: 27280304 PMCID: PMC5011188 DOI: 10.2174/1573403x12666160606121858] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Revised: 12/18/2015] [Accepted: 12/31/1969] [Indexed: 12/12/2022] Open
Abstract
The pathogenesis of cardiomyopathy and heart failure (HF) is underpinned by complex changes at subcellular, cellular and extracellular levels in the ventricular myocardium. For all of the gains that conventional treatments for HF have brought to mortality and morbidity, they do not adequately address the loss of cardiomyocyte numbers in the remodeling ventricle. Originally conceived to address this problem, cellular transplantation for HF has already gone through several stages of evolution over the past two decades. Various cell types and delivery routes have been implemented to positive effect in preclinical models of ischemic and nonischemic cardiomyopathy, with pleiotropic benefits observed in terms of myocardial remodeling, systolic and diastolic performance, perfusion, fibrosis, inflammation, metabolism and electrophysiology. To a large extent, these salubrious effects are now attributed to the indirect, paracrine capacity of transplanted stem cells to facilitate endogenous cardiac repair processes. Promising results have also followed in early phase human studies, although these have been relatively modest and somewhat inconsistent. This review details the preclinical and clinical evidence currently available regarding the use of pluripotent stem cells and adult-derived progenitor cells for cardiomyopathy and HF. It outlines the important lessons that have been learned to this point in time, and balances the promise of this exciting field against the key challenges and questions that still need to be addressed at all levels of research, to ensure that cell therapy realizes its full potential by adding to the armamentarium of HF management.
Collapse
Affiliation(s)
- Peter J Psaltis
- Co-Director of Vascular Research Centre, Heart Health Theme, South Australian Health and Medical Research Institute, North Terrace, Adelaide, South Australia, Australia 5000.
| | | | | | | |
Collapse
|
31
|
Fanton Y, Houbrechts C, Willems L, Daniëls A, Linsen L, Ratajczak J, Bronckaers A, Lambrichts I, Declercq J, Rummens JL, Hendrikx M, Hensen K. Cardiac atrial appendage stem cells promote angiogenesis in vitro and in vivo. J Mol Cell Cardiol 2016; 97:235-44. [PMID: 27291064 DOI: 10.1016/j.yjmcc.2016.06.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Revised: 05/17/2016] [Accepted: 06/08/2016] [Indexed: 12/23/2022]
Abstract
Cardiac atrial appendage stem cells (CASCs) show extraordinary myocardial differentiation properties, making them ideal candidates for myocardial regeneration. However, since the myocardium is a highly vascularized tissue, revascularization of the ischemic infarct area is essential for functional repair. Therefore, this study assessed if CASCs contribute to cardiac angiogenesis via paracrine mechanisms. First, it was demonstrated that CASCs produce and secrete high levels of numerous angiogenic growth factors, including vascular endothelial growth factor (VEGF), endothelin-1 (ET-1) and insulin-like growth factor binding protein 3 (IGFBP-3). Functional in vitro assays with a human microvascular endothelial cell line (HMEC-1) and CASC CM showed that CASCs promote endothelial cell proliferation, migration and tube formation, the most important steps of the angiogenesis process. Addition of inhibitory antibodies against identified growth factors could significantly reduce these effects, indicating their importance in CASC-induced neovascularization. The angiogenic potential of CASCs and CASC CM was also confirmed in a chorioallantoic membrane assay, demonstrating that CASCs promote blood vessel formation in vivo. In conclusion, this study shows that CASCs not only induce myocardial repair by cardiomyogenic differentiation, but also stimulate blood vessel formation by paracrine mechanisms. The angiogenic properties of CASCs further strengthen their therapeutic potential and make them an optimal stem cell source for the treatment of ischemic heart disease.
Collapse
Affiliation(s)
- Yanick Fanton
- Laboratory of Experimental Hematology, Jessa Hospital, Hasselt, Belgium; Faculty of Medicine and Life Sciences, Hasselt University, Hasselt, Belgium.
| | - Cynthia Houbrechts
- Faculty of Medicine and Life Sciences, Hasselt University, Hasselt, Belgium
| | - Leen Willems
- Laboratory of Experimental Hematology, Jessa Hospital, Hasselt, Belgium; Faculty of Medicine and Life Sciences, Hasselt University, Hasselt, Belgium
| | - Annick Daniëls
- Laboratory of Experimental Hematology, Jessa Hospital, Hasselt, Belgium
| | - Loes Linsen
- AC Biobanking, University Hospital Leuven, Leuven, Belgium
| | | | | | - Ivo Lambrichts
- Biomedical Research Institute, Hasselt University, Hasselt, Belgium
| | - Jeroen Declercq
- Laboratory of Experimental Hematology, Jessa Hospital, Hasselt, Belgium; Faculty of Medicine and Life Sciences, Hasselt University, Hasselt, Belgium
| | - Jean-Luc Rummens
- Laboratory of Experimental Hematology, Jessa Hospital, Hasselt, Belgium; Faculty of Medicine and Life Sciences, Hasselt University, Hasselt, Belgium
| | - Marc Hendrikx
- Faculty of Medicine and Life Sciences, Hasselt University, Hasselt, Belgium; Department of Cardiothoracic Surgery, Jessa Hospital, Hasselt, Belgium
| | - Karen Hensen
- Laboratory of Experimental Hematology, Jessa Hospital, Hasselt, Belgium; Faculty of Medicine and Life Sciences, Hasselt University, Hasselt, Belgium
| |
Collapse
|
32
|
Abstract
PURPOSE OF REVIEW The aim of this review was to discuss recent advances in clinical aspects of stem cell therapy in heart failure with emphasis on patient selection, stem cell types and delivery methods. RECENT FINDINGS Several stem cell types have been considered for the treatment of patients with heart failure. In nonischemic heart failure, transplantation of CD34 cells improved myocardial performance, functional capacity and neurohumoral activation. In ischemic heart failure, cardiosphere-derived cells were shown to reduce myocardial scar burden with concomitant increase in viable tissue and regional systolic wall thickening. Both autologous and allogeneic mesenchymal stem cells were shown to be effective in improving heart function in patients with ischemic heart failure; this may represent an important step toward the development of a standardized stem cell product for widespread clinical use. SUMMARY Although trials of stem cell therapy in heart failure have shown promising results, the findings are not consistent. Given the wide spectrum of heart failure, it may be difficult to define a uniform stem cell therapy for all subsets of patients; instead, future stem cell therapeutic strategies should aim for a more personalized approach by establishing optimal stem cell type, dose and delivery method for an individual patient and disease state.
Collapse
|
33
|
Hou L, Kim JJ, Woo YJ, Huang NF. Stem cell-based therapies to promote angiogenesis in ischemic cardiovascular disease. Am J Physiol Heart Circ Physiol 2015; 310:H455-65. [PMID: 26683902 DOI: 10.1152/ajpheart.00726.2015] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Accepted: 12/09/2015] [Indexed: 12/30/2022]
Abstract
Stem cell therapy is a promising approach for the treatment of tissue ischemia associated with myocardial infarction and peripheral arterial disease. Stem and progenitor cells derived from bone marrow or from pluripotent stem cells have shown therapeutic benefit in boosting angiogenesis as well as restoring tissue function. Notably, adult stem and progenitor cells including mononuclear cells, endothelial progenitor cells, and mesenchymal stem cells have progressed into clinical trials and have shown positive benefits. In this review, we overview the major classes of stem and progenitor cells, including pluripotent stem cells, and summarize the state of the art in applying these cell types for treating myocardial infarction and peripheral arterial disease.
Collapse
Affiliation(s)
- Luqia Hou
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California; Stanford Cardiovascular Institute, Stanford University, Stanford, California; and
| | - Joseph J Kim
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California; Stanford Cardiovascular Institute, Stanford University, Stanford, California; and
| | - Y Joseph Woo
- Stanford Cardiovascular Institute, Stanford University, Stanford, California; and Department of Cardiothoracic Surgery, Stanford University, Stanford, California
| | - Ngan F Huang
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California; Stanford Cardiovascular Institute, Stanford University, Stanford, California; and Department of Cardiothoracic Surgery, Stanford University, Stanford, California
| |
Collapse
|
34
|
Afzal MR, Samanta A, Shah ZI, Jeevanantham V, Abdel-Latif A, Zuba-Surma EK, Dawn B. Adult Bone Marrow Cell Therapy for Ischemic Heart Disease: Evidence and Insights From Randomized Controlled Trials. Circ Res 2015; 117:558-75. [PMID: 26160853 DOI: 10.1161/circresaha.114.304792] [Citation(s) in RCA: 165] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Accepted: 07/07/2015] [Indexed: 12/30/2022]
Abstract
RATIONALE Notwithstanding the uncertainties about the outcomes of bone marrow cell (BMC) therapy for heart repair, further insights are critically needed to improve this promising approach. OBJECTIVE To delineate the true effect of BMC therapy for cardiac repair and gain insights for future trials through systematic review and meta-analysis of data from eligible randomized controlled trials. METHODS AND RESULTS Database searches through August 2014 identified 48 eligible randomized controlled trials (enrolling 2602 patients). Weighted mean differences for changes in left ventricular (LV) ejection fraction, infarct size, LV end-systolic volume, and LV end-diastolic volume were analyzed with random-effects meta-analysis. Compared with standard therapy, BMC transplantation improved LV ejection fraction (2.92%; 95% confidence interval, 1.91-3.92; P<0.00001), reduced infarct size (-2.25%; 95% confidence interval, -3.55 to -0.95; P=0.0007) and LV end-systolic volume (-6.37 mL; 95% confidence interval, -8.95 to -3.80; P<0.00001), and tended to reduce LV end-diastolic volume (-2.26 mL; 95% confidence interval, -4.59 to 0.07; P=0.06). Similar effects were noted when data were analyzed after excluding studies with discrepancies in reporting of outcomes. The benefits also persisted when cardiac catheterization was performed in control patients as well. Although imaging modalities partly influenced the outcomes, LV ejection fraction improved in BMC-treated patients when assessed by magnetic resonance imaging. Early (<48 hours) BMC injection after myocardial Infarction was more effective in reducing infarct size, whereas BMC injection between 3 and 10 days proved superior toward improving systolic function. A minimum of 50 million BMCs seemed to be necessary, with limited additional benefits seen with increasing cell numbers. BMC therapy was safe and improved clinical outcomes, including all-cause mortality, recurrent myocardial Infarction, ventricular arrhythmia, and cerebrovascular accident during follow-up, albeit with differences between acute myocardial Infarction and chronic ischemic heart disease subgroups. CONCLUSIONS Transplantation of adult BMCs improves LV ejection fraction, reduces infarct size, and ameliorates remodeling in patients with ischemic heart disease. These effects are upheld in the analyses of studies using magnetic resonance imaging and also after excluding studies with discrepant reporting of outcomes. BMC transplantation may also reduce the incidence of death, recurrent myocardial Infarction, ventricular arrhythmia, and cerebrovascular accident during follow-up.
Collapse
Affiliation(s)
- Muhammad R Afzal
- From the Division of Cardiovascular Diseases, Cardiovascular Research Institute, and the Midwest Stem Cell Therapy Center, University of Kansas Medical Center, Kansas City (M.R.A., A.S., Z.I.S., B.D.); Heart and Vascular Specialists of Oklahoma, Oklahoma City (V.J.); Division of Cardiology, University of Kentucky, Lexington (A.A.-L.); and Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland (E.K.Z.-S)
| | - Anweshan Samanta
- From the Division of Cardiovascular Diseases, Cardiovascular Research Institute, and the Midwest Stem Cell Therapy Center, University of Kansas Medical Center, Kansas City (M.R.A., A.S., Z.I.S., B.D.); Heart and Vascular Specialists of Oklahoma, Oklahoma City (V.J.); Division of Cardiology, University of Kentucky, Lexington (A.A.-L.); and Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland (E.K.Z.-S)
| | - Zubair I Shah
- From the Division of Cardiovascular Diseases, Cardiovascular Research Institute, and the Midwest Stem Cell Therapy Center, University of Kansas Medical Center, Kansas City (M.R.A., A.S., Z.I.S., B.D.); Heart and Vascular Specialists of Oklahoma, Oklahoma City (V.J.); Division of Cardiology, University of Kentucky, Lexington (A.A.-L.); and Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland (E.K.Z.-S)
| | - Vinodh Jeevanantham
- From the Division of Cardiovascular Diseases, Cardiovascular Research Institute, and the Midwest Stem Cell Therapy Center, University of Kansas Medical Center, Kansas City (M.R.A., A.S., Z.I.S., B.D.); Heart and Vascular Specialists of Oklahoma, Oklahoma City (V.J.); Division of Cardiology, University of Kentucky, Lexington (A.A.-L.); and Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland (E.K.Z.-S)
| | - Ahmed Abdel-Latif
- From the Division of Cardiovascular Diseases, Cardiovascular Research Institute, and the Midwest Stem Cell Therapy Center, University of Kansas Medical Center, Kansas City (M.R.A., A.S., Z.I.S., B.D.); Heart and Vascular Specialists of Oklahoma, Oklahoma City (V.J.); Division of Cardiology, University of Kentucky, Lexington (A.A.-L.); and Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland (E.K.Z.-S)
| | - Ewa K Zuba-Surma
- From the Division of Cardiovascular Diseases, Cardiovascular Research Institute, and the Midwest Stem Cell Therapy Center, University of Kansas Medical Center, Kansas City (M.R.A., A.S., Z.I.S., B.D.); Heart and Vascular Specialists of Oklahoma, Oklahoma City (V.J.); Division of Cardiology, University of Kentucky, Lexington (A.A.-L.); and Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland (E.K.Z.-S)
| | - Buddhadeb Dawn
- From the Division of Cardiovascular Diseases, Cardiovascular Research Institute, and the Midwest Stem Cell Therapy Center, University of Kansas Medical Center, Kansas City (M.R.A., A.S., Z.I.S., B.D.); Heart and Vascular Specialists of Oklahoma, Oklahoma City (V.J.); Division of Cardiology, University of Kentucky, Lexington (A.A.-L.); and Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland (E.K.Z.-S).
| |
Collapse
|
35
|
Fisher SA, Doree C, Mathur A, Martin-Rendon E. Meta-Analysis of Cell Therapy Trials for Patients With Heart Failure. Circ Res 2015; 116:1361-77. [DOI: 10.1161/circresaha.116.304386] [Citation(s) in RCA: 181] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2014] [Accepted: 01/20/2015] [Indexed: 02/06/2023]
Affiliation(s)
- Sheila A Fisher
- From the Systematic Review Group, R&D Department, NHS Blood and Transplant, Oxford, UK (S.A.F., C.D.); Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, Oxford, UK (S.A.F., C.D., E.M.-R.); Department of Clinical Pharmacology, William Harvey Research Institute, Queen Mary University of London, London, UK (A.M.); and Stem Cell Research Laboratory, R&D Department, NHS Blood and Transplant, Oxford, UK (E.M.-R.)
| | - Carolyn Doree
- From the Systematic Review Group, R&D Department, NHS Blood and Transplant, Oxford, UK (S.A.F., C.D.); Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, Oxford, UK (S.A.F., C.D., E.M.-R.); Department of Clinical Pharmacology, William Harvey Research Institute, Queen Mary University of London, London, UK (A.M.); and Stem Cell Research Laboratory, R&D Department, NHS Blood and Transplant, Oxford, UK (E.M.-R.)
| | - Anthony Mathur
- From the Systematic Review Group, R&D Department, NHS Blood and Transplant, Oxford, UK (S.A.F., C.D.); Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, Oxford, UK (S.A.F., C.D., E.M.-R.); Department of Clinical Pharmacology, William Harvey Research Institute, Queen Mary University of London, London, UK (A.M.); and Stem Cell Research Laboratory, R&D Department, NHS Blood and Transplant, Oxford, UK (E.M.-R.)
| | - Enca Martin-Rendon
- From the Systematic Review Group, R&D Department, NHS Blood and Transplant, Oxford, UK (S.A.F., C.D.); Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, Oxford, UK (S.A.F., C.D., E.M.-R.); Department of Clinical Pharmacology, William Harvey Research Institute, Queen Mary University of London, London, UK (A.M.); and Stem Cell Research Laboratory, R&D Department, NHS Blood and Transplant, Oxford, UK (E.M.-R.)
| |
Collapse
|
36
|
Almeida SO, Skelton RJ, Adigopula S, Ardehali R. Arrhythmia in stem cell transplantation. Card Electrophysiol Clin 2015; 7:357-70. [PMID: 26002399 DOI: 10.1016/j.ccep.2015.03.012] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Stem cell regenerative therapies hold promise for treating diseases across the spectrum of medicine. While significant progress has been made in the preclinical stages, the clinical application of cardiac cell therapy is limited by technical challenges. Certain methods of cell delivery, such as intramyocardial injection, carry a higher rate of arrhythmias. Other potential contributors to the arrhythmogenicity of cell transplantation include reentrant pathways caused by heterogeneity in conduction velocities between graft and host as well as graft automaticity. In this article, the arrhythmogenic potential of cell delivery to the heart is discussed.
Collapse
Affiliation(s)
- Shone O Almeida
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, 100 UCLA Medical Plaza, Suite 630 East, Los Angeles, CA 90095, USA
| | - Rhys J Skelton
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, 100 UCLA Medical Plaza, Suite 630 East, Los Angeles, CA 90095, USA; Murdoch Children's Research Institute, The Royal Children's Hospital, Cardiac Development, 50 Flemington Road, Parkville, Victoria 3052, Australia
| | - Sasikanth Adigopula
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, 100 UCLA Medical Plaza, Suite 630 East, Los Angeles, CA 90095, USA
| | - Reza Ardehali
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, 100 UCLA Medical Plaza, Suite 630 East, Los Angeles, CA 90095, USA; Eli and Edyth Broad Stem Cell Research Center, University of California, 675 Charles E Young Drive South, MRL Room 3780, Los Angeles, CA 90095, USA.
| |
Collapse
|
37
|
Golpanian S, El-Khorazaty J, Mendizabal A, DiFede DL, Suncion VY, Karantalis V, Fishman JE, Ghersin E, Balkan W, Hare JM. Effect of aging on human mesenchymal stem cell therapy in ischemic cardiomyopathy patients. J Am Coll Cardiol 2015; 65:125-32. [PMID: 25593053 DOI: 10.1016/j.jacc.2014.10.040] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Revised: 09/21/2014] [Accepted: 10/14/2014] [Indexed: 12/29/2022]
Abstract
BACKGROUND The role of patient age in the efficacy of mesenchymal stem cell (MSC) therapy in ischemic cardiomyopathy (ICM) is controversial. OBJECTIVES This study sought to determine whether the therapeutic effect of culture-expanded MSCs persists, even in older subjects. METHODS Patients with ICM who received MSCs via transendocardial stem cell injection (TESI) as part of the TAC-HFT (Transendocardial Autologous Cells in Ischemic Heart Failure) (n = 19) and POSEIDON (Percutaneous Stem Cell Injection Delivery Effects on Neomyogenesis) (n = 30) clinical trials were divided into 2 age groups: younger than 60 and 60 years of age and older. Functional capacity was measured by 6-min walk distance (6MWD) and quality of life using the Minnesota Living With Heart Failure Questionnaire (MLHFQ) score, measured at baseline, 6 months, and 1 year post-TESI. Various cardiac imaging parameters, including absolute scar size, were compared at baseline and 1 year post-TESI. RESULTS The mean 6MWD was similar at baseline and increased at 1 year post-TESI in both groups: 48.5 ± 14.6 m (p = 0.001) for the younger and 35.9 ± 18.3 m (p = 0.038) for the older participants (p = NS between groups). The older group exhibited a significant reduction in MLHFQ score (-7.04 ± 3.54; p = 0.022), whereas the younger than 60 age group had a borderline significant reduction (-11.22 ± 5.24; p = 0.058) from baseline (p = NS between groups). Although there were significant reductions in absolute scar size from baseline to 1 year post-TESI, the effect did not differ by age. CONCLUSIONS MSC therapy with TESI in ICM patients improves 6MWD and MLHFQ score and reduces myocardial infarction size. Importantly, older individuals did not have an impaired response to MSC therapy.
Collapse
Affiliation(s)
- Samuel Golpanian
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida
| | | | | | - Darcy L DiFede
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida
| | - Viky Y Suncion
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida
| | - Vasileios Karantalis
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida
| | - Joel E Fishman
- Department of Radiology, University of Miami Miller School of Medicine, Miami, Florida
| | - Eduard Ghersin
- Department of Radiology, University of Miami Miller School of Medicine, Miami, Florida
| | - Wayne Balkan
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida
| | - Joshua M Hare
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida.
| |
Collapse
|
38
|
Harvey E, Fisher SA, Doree C, Taggart DP, Martin-Rendon E. Current evidence of the efficacy of cell-based therapies in heart failure. Circ J 2015; 79:229-36. [PMID: 25744736 DOI: 10.1253/circj.cj-14-1415] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Heart failure (HF) is the major cause of mortality worldwide. For more than a decade, cell-based therapies have been developed as treatment for heart disease as an alternative to current therapies. Trials and systematic reviews have assessed the safety and efficacy of cell therapies in a diverse number of participants and clinical settings. The present study collated and synthesized evidence from all systematic reviews related to cell-based therapies and HF. A total of 11 systematic reviews were identified through searches of electronic databases up to June 2014. We set out to answer 2 key questions on the efficacy of cell therapies in HF: (1) What is the overall effect of cell therapies on primary outcomes such as left ventricular ejection fraction (LVEF) and mortality? (2) How important is it to define the clinical setting and length of follow-up when assessing cell therapies and HF? There seems to be enough evidence to suggest that cell therapies have a moderate, long-lasting effect on LVEF, but the reduction on the risk of mortality observed by some systematic reviews needs to be confirmed in larger, statistically powered clinical trials. Additionally, and in order to strengthen conclusions, it is important to assess clinical evidence for defined clinical settings and to standardize the length of follow-up when comparing outcome data across several trials and systematic reviews.
Collapse
Affiliation(s)
- Emma Harvey
- Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford; Stem Cell Research Laboratory, NHS Blood and Transplant, Oxford, UK
| | | | | | | | | |
Collapse
|
39
|
Cantero Peral S, Burkhart HM, Oommen S, Yamada S, Nyberg SL, Li X, O'Leary PW, Terzic A, Cannon BC, Nelson TJ. Safety and feasibility for pediatric cardiac regeneration using epicardial delivery of autologous umbilical cord blood-derived mononuclear cells established in a porcine model system. Stem Cells Transl Med 2015; 4:195-206. [PMID: 25561683 DOI: 10.5966/sctm.2014-0195] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Congenital heart diseases (CHDs) requiring surgical palliation mandate new treatment strategies to optimize long-term outcomes. Despite the mounting evidence of cardiac regeneration, there are no long-term safety studies of autologous cell-based transplantation in the pediatric setting. We aimed to establish a porcine pipeline to evaluate the feasibility and long-term safety of autologous umbilical cord blood mononuclear cells (UCB-MNCs) transplanted into the right ventricle (RV) of juvenile porcine hearts. Piglets were born by caesarean section to enable UCB collection. Upon meeting release criteria, 12 animals were randomized in a double-blinded fashion prior to surgical delivery of test article (n=6) or placebo (n=6). The UCB-MNC (3×10(6) cells per kilogram) or control (dimethyl sulfoxide, 10%) products were injected intramyocardially into the RV under direct visualization. The cohorts were monitored for 3 months after product delivery with assessments of cardiac performance, rhythm, and serial cardiac biochemical markers, followed by terminal necropsy. No mortalities were associated with intramyocardial delivery of UCB-MNCs or placebo. Two animals from the placebo group developed local skin infection after surgery that responded to antibiotic treatment. Electrophysiological assessments revealed no arrhythmias in either group throughout the 3-month study. Two animals in the cell-therapy group had transient, subclinical dysrhythmia in the perioperative period, likely because of an exaggerated response to anesthesia. Overall, this study demonstrated that autologous UCB-MNCs can be safely collected and surgically delivered in a pediatric setting. The safety profile establishes the foundation for cell-based therapy directed at the RV of juvenile hearts and aims to accelerate cell-based therapies toward clinical trials for CHD.
Collapse
Affiliation(s)
- Susana Cantero Peral
- Division of General Internal Medicine, Center for Regenerative Medicine, Pediatric Cardiothoracic Surgery, Division of Cardiovascular Diseases, Transplant Center, Division of Biomedical Statistics and Informatics, Division of Pediatric Cardiology, Department of Molecular Pharmacology and Experimental Therapeutics, and Mayo Clinic, Rochester, Minnesota, USA; Program of Doctorate of Internal Medicine, Autonomous University of Barcelona, Barcelona, Spain
| | - Harold M Burkhart
- Division of General Internal Medicine, Center for Regenerative Medicine, Pediatric Cardiothoracic Surgery, Division of Cardiovascular Diseases, Transplant Center, Division of Biomedical Statistics and Informatics, Division of Pediatric Cardiology, Department of Molecular Pharmacology and Experimental Therapeutics, and Mayo Clinic, Rochester, Minnesota, USA; Program of Doctorate of Internal Medicine, Autonomous University of Barcelona, Barcelona, Spain
| | - Saji Oommen
- Division of General Internal Medicine, Center for Regenerative Medicine, Pediatric Cardiothoracic Surgery, Division of Cardiovascular Diseases, Transplant Center, Division of Biomedical Statistics and Informatics, Division of Pediatric Cardiology, Department of Molecular Pharmacology and Experimental Therapeutics, and Mayo Clinic, Rochester, Minnesota, USA; Program of Doctorate of Internal Medicine, Autonomous University of Barcelona, Barcelona, Spain
| | - Satsuki Yamada
- Division of General Internal Medicine, Center for Regenerative Medicine, Pediatric Cardiothoracic Surgery, Division of Cardiovascular Diseases, Transplant Center, Division of Biomedical Statistics and Informatics, Division of Pediatric Cardiology, Department of Molecular Pharmacology and Experimental Therapeutics, and Mayo Clinic, Rochester, Minnesota, USA; Program of Doctorate of Internal Medicine, Autonomous University of Barcelona, Barcelona, Spain
| | - Scott L Nyberg
- Division of General Internal Medicine, Center for Regenerative Medicine, Pediatric Cardiothoracic Surgery, Division of Cardiovascular Diseases, Transplant Center, Division of Biomedical Statistics and Informatics, Division of Pediatric Cardiology, Department of Molecular Pharmacology and Experimental Therapeutics, and Mayo Clinic, Rochester, Minnesota, USA; Program of Doctorate of Internal Medicine, Autonomous University of Barcelona, Barcelona, Spain
| | - Xing Li
- Division of General Internal Medicine, Center for Regenerative Medicine, Pediatric Cardiothoracic Surgery, Division of Cardiovascular Diseases, Transplant Center, Division of Biomedical Statistics and Informatics, Division of Pediatric Cardiology, Department of Molecular Pharmacology and Experimental Therapeutics, and Mayo Clinic, Rochester, Minnesota, USA; Program of Doctorate of Internal Medicine, Autonomous University of Barcelona, Barcelona, Spain
| | - Patrick W O'Leary
- Division of General Internal Medicine, Center for Regenerative Medicine, Pediatric Cardiothoracic Surgery, Division of Cardiovascular Diseases, Transplant Center, Division of Biomedical Statistics and Informatics, Division of Pediatric Cardiology, Department of Molecular Pharmacology and Experimental Therapeutics, and Mayo Clinic, Rochester, Minnesota, USA; Program of Doctorate of Internal Medicine, Autonomous University of Barcelona, Barcelona, Spain
| | - Andre Terzic
- Division of General Internal Medicine, Center for Regenerative Medicine, Pediatric Cardiothoracic Surgery, Division of Cardiovascular Diseases, Transplant Center, Division of Biomedical Statistics and Informatics, Division of Pediatric Cardiology, Department of Molecular Pharmacology and Experimental Therapeutics, and Mayo Clinic, Rochester, Minnesota, USA; Program of Doctorate of Internal Medicine, Autonomous University of Barcelona, Barcelona, Spain
| | - Bryan C Cannon
- Division of General Internal Medicine, Center for Regenerative Medicine, Pediatric Cardiothoracic Surgery, Division of Cardiovascular Diseases, Transplant Center, Division of Biomedical Statistics and Informatics, Division of Pediatric Cardiology, Department of Molecular Pharmacology and Experimental Therapeutics, and Mayo Clinic, Rochester, Minnesota, USA; Program of Doctorate of Internal Medicine, Autonomous University of Barcelona, Barcelona, Spain
| | - Timothy J Nelson
- Division of General Internal Medicine, Center for Regenerative Medicine, Pediatric Cardiothoracic Surgery, Division of Cardiovascular Diseases, Transplant Center, Division of Biomedical Statistics and Informatics, Division of Pediatric Cardiology, Department of Molecular Pharmacology and Experimental Therapeutics, and Mayo Clinic, Rochester, Minnesota, USA; Program of Doctorate of Internal Medicine, Autonomous University of Barcelona, Barcelona, Spain
| |
Collapse
|
40
|
Indumathi S, Harikrishnan R, Dhanasekaran M. Redundant Human Omentum Fat: A Leap Towards Regenerative Medicine. Regen Med 2015. [DOI: 10.1007/978-1-4471-6542-2_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
|
41
|
Wang H, Wang Z, Jiang H, Ma D, Zhou W, Zhang G, Chen W, Huang J, Liu Y. Effect of autologous bone marrow cell transplantation combined with off-pump coronary artery bypass grafting on cardiac function in patients with chronic myocardial infarction. Cardiology 2014; 130:27-33. [PMID: 25501100 DOI: 10.1159/000369381] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Accepted: 10/24/2014] [Indexed: 11/19/2022]
Abstract
OBJECTIVES This study aimed to investigate the feasibility and effects of intramuscular injections of autologous bone marrow cells (BMC) combined with off-pump coronary artery bypass grafts (OPCAB) on improving cardiac function in chronic myocardial infarction patients. METHODS Ninety patients with chronic myocardial infarction were prospectively enrolled and randomized to an OPCAB with saline or an OPCAB with BMC-treatment group. After finishing CABG, patients received injections of BMC or saline into the marginal area of the infarct. The primary endpoint was incidence of emergent adverse events within 6 months. RESULTS There were no differences between the control and BMC-treated groups in baseline ejection fractions (EF) or wall motion score indices (WMSI) in the affected segments. At the 6-month follow-up, the ejection fraction was significantly increased in the BMC-treated group compared to controls (47.58 ± 6.34 vs. 40.11 ± 7.42; p < 0.05), whereas the WMSI were significantly decreased (1.25 ± 0.32 vs. 1.54 ± 0.53; p < 0.05), with no occurrences of life-threatening arrhythmias or death. The addition of BMC injections to OPCAB treatment increased regional perfusion to the marginal infarct area. CONCLUSION These results demonstrate that BMC transplant is beneficial to the cardiac function with no adverse effects, and therefore a safe and feasible adjunct therapy providing beneficial effects in clinical practice.
Collapse
Affiliation(s)
- Huishan Wang
- Department of Cardiac Surgery, Shenyang Northern Hospital, Shenyang, PR China
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Madonna R, Ferdinandy P, De Caterina R, Willerson JT, Marian AJ. Recent developments in cardiovascular stem cells. Circ Res 2014; 115:e71-8. [PMID: 25477490 DOI: 10.1161/circresaha.114.305567] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Rosalinda Madonna
- From the Center of Excellence on Aging, Institute of Cardiology, Department of Neuroscience and Imaging, "G. d'Annunzio" University, Chieti, Italy (R.M., R.D.C.); Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Texas Heart Institute, Houston (R.M., J.T.W.); Division of Cardiology, Department of Internal Medicine (R.M., J.T.W., A.J.M.), and Institute of Molecular Medicine, The University of Texas Health Science Center, Houston (A.J.M.)
| | - Peter Ferdinandy
- From the Center of Excellence on Aging, Institute of Cardiology, Department of Neuroscience and Imaging, "G. d'Annunzio" University, Chieti, Italy (R.M., R.D.C.); Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Texas Heart Institute, Houston (R.M., J.T.W.); Division of Cardiology, Department of Internal Medicine (R.M., J.T.W., A.J.M.), and Institute of Molecular Medicine, The University of Texas Health Science Center, Houston (A.J.M.)
| | - Raffaele De Caterina
- From the Center of Excellence on Aging, Institute of Cardiology, Department of Neuroscience and Imaging, "G. d'Annunzio" University, Chieti, Italy (R.M., R.D.C.); Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Texas Heart Institute, Houston (R.M., J.T.W.); Division of Cardiology, Department of Internal Medicine (R.M., J.T.W., A.J.M.), and Institute of Molecular Medicine, The University of Texas Health Science Center, Houston (A.J.M.)
| | - James T Willerson
- From the Center of Excellence on Aging, Institute of Cardiology, Department of Neuroscience and Imaging, "G. d'Annunzio" University, Chieti, Italy (R.M., R.D.C.); Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Texas Heart Institute, Houston (R.M., J.T.W.); Division of Cardiology, Department of Internal Medicine (R.M., J.T.W., A.J.M.), and Institute of Molecular Medicine, The University of Texas Health Science Center, Houston (A.J.M.)
| | - Ali J Marian
- From the Center of Excellence on Aging, Institute of Cardiology, Department of Neuroscience and Imaging, "G. d'Annunzio" University, Chieti, Italy (R.M., R.D.C.); Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Texas Heart Institute, Houston (R.M., J.T.W.); Division of Cardiology, Department of Internal Medicine (R.M., J.T.W., A.J.M.), and Institute of Molecular Medicine, The University of Texas Health Science Center, Houston (A.J.M.).
| |
Collapse
|
43
|
Sharp TE, George JC. Stem cell therapy and breast cancer treatment: review of stem cell research and potential therapeutic impact against cardiotoxicities due to breast cancer treatment. Front Oncol 2014; 4:299. [PMID: 25405100 PMCID: PMC4217360 DOI: 10.3389/fonc.2014.00299] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Accepted: 10/14/2014] [Indexed: 12/16/2022] Open
Abstract
A new problem has emerged with the ever-increasing number of breast cancer survivors. While early screening and advances in treatment have allowed these patients to overcome their cancer, these treatments often have adverse cardiovascular side effects that can produce abnormal cardiovascular function. Chemotherapeutic and radiation therapy have both been linked to cardiotoxicity; these therapeutics can cause a loss of cardiac muscle and deterioration of vascular structure that can eventually lead to heart failure (HF). This cardiomyocyte toxicity can leave the breast cancer survivor with a probable diagnosis of dilated or restrictive cardiomyopathy (DCM or RCM). While current HF standard of care can alleviate symptoms, other than heart transplantation, there is no therapy that replaces cardiac myocytes that are killed during cancer therapies. There is a need to develop novel therapeutics that can either prevent or reverse the cardiac injury caused by cancer therapeutics. These new therapeutics should promote the regeneration of lost or deteriorating myocardium. Over the last several decades, the therapeutic potential of cell-based therapy has been investigated for HF patients. In this review, we discuss the progress of pre-clinical and clinical stem cell research for the diseased heart and discuss the possibility of utilizing these novel therapies to combat cardiotoxicity observed in breast cancer survivors.
Collapse
Affiliation(s)
- Thomas E Sharp
- Cardiovascular Research Center, Temple University School of Medicine , Philadelphia, PA , USA
| | - Jon C George
- Cardiovascular Research Center, Temple University School of Medicine , Philadelphia, PA , USA ; Division of Cardiovascular Medicine, Temple University Hospital , Philadelphia, PA , USA
| |
Collapse
|
44
|
Chong JJH, Murry CE. Cardiac regeneration using pluripotent stem cells--progression to large animal models. Stem Cell Res 2014; 13:654-65. [PMID: 25087896 PMCID: PMC4253057 DOI: 10.1016/j.scr.2014.06.005] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Revised: 06/18/2014] [Accepted: 06/28/2014] [Indexed: 12/23/2022] Open
Abstract
Pluripotent stem cells (PSCs) have indisputable cardiomyogenic potential and therefore have been intensively investigated as a potential cardiac regenerative therapy. Current directed differentiation protocols are able to produce high yields of cardiomyocytes from PSCs and studies in small animal models of cardiovascular disease have proven sustained engraftment and functional efficacy. Therefore, the time is ripe for cardiac regenerative therapies using PSC derivatives to be tested in large animal models that more closely resemble the hearts of humans. In this review, we discuss the results of our recent study using human embryonic stem cell derived cardiomyocytes (hESC-CM) in a non-human primate model of ischemic cardiac injury. Large scale remuscularization, electromechanical coupling and short-term arrhythmias demonstrated by our hESC-CM grafts are discussed in the context of other studies using adult stem cells for cardiac regeneration.
Collapse
Affiliation(s)
- James J H Chong
- Department of Cardiology Westmead Hospital, Sydney, NSW, Australia; School of Medicine, University of Sydney, Sydney, NSW, Australia; Westmead Millennium Institute for Medical Research, Sydney, NSW, Australia; Center for Cardiovascular Biology, University of Washington, Seattle, WA, USA; Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA.
| | - Charles E Murry
- Center for Cardiovascular Biology, University of Washington, Seattle, WA, USA; Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA; Department of Pathology, University of Washington, Seattle, WA, USA; Department of Medicine/Cardiology, University of Washington, Seattle, WA, USA; Department of Bioengineering, University of Washington, Seattle, WA, USA
| |
Collapse
|
45
|
The Current State of Stem Cell Therapeutics: Canadian Approaches in the International Context. Can J Cardiol 2014; 30:1361-9. [DOI: 10.1016/j.cjca.2014.04.031] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2014] [Revised: 04/24/2014] [Accepted: 04/27/2014] [Indexed: 11/22/2022] Open
|
46
|
Ali-Hasan-Al-Saegh S, Mirhosseini SJ, Lotfaliani MR, Dehghan HR, Sedaghat-Hamedani F, Kayvanpour E, Rezaeisadrabadi M, Ghaffari N, Vahabzadeh V, Jebran AF, Sabashnikov A, Popov AF. Transplantation of bone marrow stem cells during cardiac surgery. Asian Cardiovasc Thorac Ann 2014; 23:363-74. [PMID: 25281762 DOI: 10.1177/0218492314553251] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
BACKGROUND This systematic review with meta-analysis sought to determine the efficacy and safety of intramyocardial transplantation of bone marrow stem cells during coronary artery bypass graft surgery on postoperative cardiac functional parameters such as left ventricular ejection fraction and left ventricular end-diastolic volume. METHODS Medline/PubMed, Embase, Elsevier, Sciences online database, and Google Scholar literature search were searched. The effect sizes measured were risk ratio for categorical variables and weighted mean difference with 95% confidence interval for calculating differences between mean values of baseline and follow-up cardiac functional parameters. A value of p < 0.1 for Q test, or I(2 )> 50%, indicated significant heterogeneity among studies. The literature search retrieved 2900 studies from screened databases, of which 2866 (98.6%) were excluded and 34 (619 patients) were included for scoping review. The final analysis included 9 studies (335 patients). RESULTS Pooled effects estimates of left ventricular ejection fraction and left ventricular end-diastolic volume showed that bone marrow stem cell transplantation had a weighted mean difference of 4.06 (95% confidence interval: 0.41-7.72; p = 0.02) and 7.06 (95% confidence interval: -8.58-22.7; p = 0.3), respectively. CONCLUSIONS Intramyocardial transplantation of bone marrow stem cells improves cardiac functional parameters, significantly increasing left ventricular ejection fraction with a nonsignificant reduction in left ventricular end-diastolic volume. Also, this therapeutic method has no life-threatening complications and was therefore found to be an effective and safe method.
Collapse
Affiliation(s)
- Sadeq Ali-Hasan-Al-Saegh
- Cardiovascular Research Center, Afshar Hospital, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Seyed Jalil Mirhosseini
- Cardiovascular Research Center, Afshar Hospital, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Mohammad-Reza Lotfaliani
- Cardiovascular Research Center, Afshar Hospital, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Hamid Reza Dehghan
- Cardiovascular Research Center, Afshar Hospital, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | | | - Elham Kayvanpour
- Department of Medicine III, University of Heidelberg, Heidelberg, Germany
| | - Mohammad Rezaeisadrabadi
- Cardiovascular Research Center, Afshar Hospital, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Naser Ghaffari
- Department of Cardiovascular Surgery, Herzchirurgie Klinikum, Karlsruhe, Germany
| | - Vahid Vahabzadeh
- Department of Cardiovascular Surgery, Herzchirurgie Klinikum, Karlsruhe, Germany
| | - Ahamd Fawad Jebran
- Department of Thoracic and Cardiovascular Surgery, University Hospital Goettingen, Goettingen, Germany
| | - Anton Sabashnikov
- Department of Cardiothoracic Transplantation and Mechanical Circulatory Support, Royal Brompton & Harefield NHS Foundation Trust, London, UK
| | - Aron-Frederik Popov
- Department of Cardiothoracic Transplantation and Mechanical Circulatory Support, Royal Brompton & Harefield NHS Foundation Trust, London, UK
| |
Collapse
|
47
|
Xiao C, Zhou S, Liu Y, Hu H. Efficacy and safety of bone marrow cell transplantation for chronic ischemic heart disease: a meta-analysis. Med Sci Monit 2014; 20:1768-77. [PMID: 25270584 PMCID: PMC4199404 DOI: 10.12659/msm.892047] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Background Although bone marrow-derived cells (BMCs) have shown great therapeutic potential in patients with chronic ischemic heart disease (CIHD), the exact efficacy and safety of BMCs therapy is still not completely defined. Material/Methods We searched PubMed, OVID, EMBASE, the Cochrane Library, and ClinicalTrials.gov and finally identified 20 qualified trials in this meta-analysis. Assessment of efficacy was based on left ventricular ejection fraction (LVEF), left ventricular end-systolic volume (LVESV), and left ventricular end-diastolic volume (LVEDV) improvement, by weighted mean difference (WMD) with 95% confidence intervals (CIs). Results of all-cause death, ventricular arrhythmia, recurrent myocardial infarction, and cerebrovascular accident were pooled to assess safety. Subgroup analysis was performed by stratifying RCTs into 2 subgroups of those with revascularization and without revascularization. Results BMC transplantation significantly improved LVEF in patients with revascularization (3.35%, 95% CI 0.72% to 5.97%, p=0.01; I2=85%) and without revascularization (3.05%, 95% CI 0.65% to 5.45%, p=0.01; I2=86%). In patients without revascularization, BMC transplantation was associated with significantly decreased LVESV (−11.75 ml, 95% CI −17.81 ml to −5.69 ml, p=0.0001; I2=81%), and LVEDV (−7.80 ml, 95% CI −15.31 ml to −0.29 ml, p=0.04; I2=39%). Subgroup analysis showed that the route of transplantation, baseline LVEF, and type of cells delivered could influence the efficacy of BMC transplantation. Conclusions Autologous transplantation of BMCs was safe and effective for patients who were candidates for revascularization with CABG/PCI and those who were not. However, large clinical trials and long-term follow-up are required to confirm these benefits.
Collapse
Affiliation(s)
- Chun Xiao
- Department of Medicine, Sichuan Scientific & Cellular Biotechnology Research Institute, Chengdu, China (mainland)
| | - Shijie Zhou
- Department of Medicine, Sichuan Scientific & Cellular Biotechnology Research Institute, Chengdu, China (mainland)
| | - Yueqiang Liu
- Department of Medicine, Sichuan Scientific & Cellular Biotechnology Research Institute, Chengdu, China (mainland)
| | - Huozhen Hu
- Department of Medicine, Sichuan Scientific & Cellular Biotechnology Research Institute, Chengdu, China (mainland)
| |
Collapse
|
48
|
Autologous bone marrow mononuclear cell transplantation in ischemic heart failure: A prospective, controlled, randomized, double-blind study of cell transplantation combined with coronary bypass. J Heart Lung Transplant 2014; 33:567-74. [DOI: 10.1016/j.healun.2014.02.009] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Revised: 11/23/2013] [Accepted: 02/07/2014] [Indexed: 02/06/2023] Open
|
49
|
Fisher SA, Brunskill SJ, Doree C, Mathur A, Taggart DP, Martin-Rendon E. Stem cell therapy for chronic ischaemic heart disease and congestive heart failure. Cochrane Database Syst Rev 2014:CD007888. [PMID: 24777540 DOI: 10.1002/14651858.cd007888.pub2] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
BACKGROUND A promising approach to the treatment of chronic ischaemic heart disease (IHD) and heart failure is the use of stem cells. The last decade has seen a plethora of randomised controlled trials (RCTs) developed worldwide which have generated conflicting results. OBJECTIVES The critical evaluation of clinical evidence on the safety and efficacy of autologous adult bone marrow-derived stem cells (BMSC) as a treatment for chronic ischaemic heart disease (IHD) and heart failure. SEARCH METHODS We searched the Cochrane Central Register of Controlled Trials (CENTRAL) (The Cochrane Library, 2013, Issue 3), MEDLINE (from 1950), EMBASE (from 1974), CINAHL (from 1982) and the Transfusion Evidence Library (from 1980), together with ongoing trial databases, for relevant trials up to 31st March 2013. SELECTION CRITERIA Eligible studies included RCTs comparing autologous adult stem/progenitor cells with no autologous stem/progenitor cells in participants with chronic IHD and heart failure. Co-interventions such as primary angioplasty, surgery or administration of stem cell mobilising agents, were included where administered to treatment and control arms equally. DATA COLLECTION AND ANALYSIS Two review authors independently screened all references for eligibility, assessed trial quality and extracted data. We undertook a quantitative evaluation of data using fixed-effect meta-analyses. We evaluated heterogeneity using the I² statistic; we explored considerable heterogeneity (I² > 75%) using a random-effects model and subgroup analyses. MAIN RESULTS We include 23 RCTs involving 1255 participants in this review. Risk of bias was generally low, with the majority of studies reporting appropriate methods of randomisation and blinding, Autologous bone marrow stem cell treatment reduced the incidence of mortality (risk ratio (RR) 0.28, 95% confidence interval (CI) 0.14 to 0.53, P = 0.0001, 8 studies, 494 participants, low quality evidence) and rehospitalisation due to heart failure (RR 0.26, 95% CI 0.07 to 0.94, P = 0.04, 2 studies, 198 participants, low quality evidence) in the long term (≥12 months). The treatment had no clear effect on mortality (RR 0.68, 95% CI 0.32 to 1.41, P = 0.30, 21 studies, 1138 participants, low quality evidence) or rehospitalisation due to heart failure (RR 0.36, 95% CI 0.12 to 1.06, P = 0.06, 4 studies, 236 participants, low quality evidence) in the short term (< 12 months), which is compatible with benefit, no difference or harm. The treatment was also associated with a reduction in left ventricular end systolic volume (LVESV) (mean difference (MD) -14.64 ml, 95% CI -20.88 ml to -8.39 ml, P < 0.00001, 3 studies, 153 participants, moderate quality evidence) and stroke volume index (MD 6.52, 95% CI 1.51 to 11.54, P = 0.01, 2 studies, 62 participants, moderate quality evidence), and an improvement in left ventricular ejection fraction (LVEF) (MD 2.62%, 95% CI 0.50% to 4.73%, P = 0.02, 6 studies, 254 participants, moderate quality evidence), all at long-term follow-up. Overall, we observed a reduction in functional class (New York Heart Association (NYHA) class) in favour of BMSC treatment during short-term follow-up (MD -0.63, 95% CI -1.08 to -0.19, P = 0.005, 11 studies, 486 participants, moderate quality evidence) and long-term follow-up (MD -0.91, 95% CI -1.38 to -0.44, P = 0.0002, 4 studies, 196 participants, moderate quality evidence), as well as a difference in Canadian Cardiovascular Society score in favour of BMSC (MD -0.81, 95% CI -1.55 to -0.07, P = 0.03, 8 studies, 379 participants, moderate quality evidence). Of 19 trials in which adverse events were reported, adverse events relating to the BMSC treatment or procedure occurred in only four individuals. No long-term adverse events were reported. Subgroup analyses conducted for outcomes such as LVEF and NYHA class revealed that (i) route of administration, (ii) baseline LVEF, (iii) cell type, and (iv) clinical condition are important factors that may influence treatment effect. AUTHORS' CONCLUSIONS This systematic review and meta-analysis found moderate quality evidence that BMSC treatment improves LVEF. Unlike in trials where BMSC were administered following acute myocardial infarction (AMI), we found some evidence for a potential beneficial clinical effect in terms of mortality and performance status in the long term (after at least one year) in people who suffer from chronic IHD and heart failure, although the quality of evidence was low.
Collapse
Affiliation(s)
- Sheila A Fisher
- Systematic Review Initiative, NHS Blood and Transplant, Level 2, John Radcliffe Hospital, Headington, Oxford, Oxon, UK, OX3 9BQ
| | | | | | | | | | | |
Collapse
|
50
|
Greiner JFW, Grunwald LM, Müller J, Sudhoff H, Widera D, Kaltschmidt C, Kaltschmidt B. Culture bag systems for clinical applications of adult human neural crest-derived stem cells. Stem Cell Res Ther 2014; 5:34. [PMID: 24629140 PMCID: PMC4055128 DOI: 10.1186/scrt422] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Accepted: 03/11/2014] [Indexed: 11/15/2022] Open
Abstract
Introduction Facing the challenging treatment of neurodegenerative diseases as well as complex craniofacial injuries such as those common after cancer therapy, the field of regenerative medicine increasingly relies on stem cell transplantation strategies. Here, neural crest-derived stem cells (NCSCs) offer many promising applications, although scale up of clinical-grade processes prior to potential transplantations is currently limiting. In this study, we aimed to establish a clinical-grade, cost-reducing cultivation system for NCSCs isolated from the adult human nose using cGMP-grade Afc-FEP bags. Methods We cultivated human neural crest-derived stem cells from inferior turbinate (ITSCs) in a cell culture bag system using Afc-FEP bags in human blood plasma-supplemented medium. Investigations of viability, proliferation and expression profile of bag-cultured ITSCs were followed by DNA-content and telomerase activity determination. Cultivated ITSCs were introduced to directed in vitro differentiation assays to assess their potential for mesodermal and ectodermal differentiation. Mesodermal differentiation was determined using an enzyme activity assay (alkaline phosphatase, ALP), respective stainings (Alizarin Red S, Von Kossa and Oil Red O), and RT-PCR, while immunocytochemistry and synaptic vesicle recycling were applied to assay neuroectodermal differentiation of ITSCs. Results When cultivated within Afc-FEP bags, ITSCs grew three-dimensionally in a human blood plasma-derived matrix, thereby showing unchanged morphology, proliferation capability, viability and expression profile in comparison to three dimensionally-cultured ITSCs growing in standard cell culture plastics. Genetic stability of bag-cultured ITSCs was further accompanied by unchanged telomerase activity. Importantly, ITSCs retained their potential to differentiate into mesodermal cell types, particularly including ALP-active, Alizarin Red S-, and Von Kossa-positive osteogenic cell types, as well as adipocytes positive in Oil Red O assays. Bag culture further did not affect the potential of ITSCs to undergo differentiation into neuroectodermal cell types coexpressing β-III-tubulin and MAP2 and exhibiting the capability for synaptic vesicle recycling. Conclusions Here, we report for the first time the successful cultivation of human NCSCs within cGMP-grade Afc-FEP bags using a human blood plasma-supplemented medium. Our findings particularly demonstrate the unchanged differentiation capability and genetic stability of the cultivated NCSCs, suggesting the great potential of this culture system for future medical applications in the field of regenerative medicine.
Collapse
|